163
Views
1
CrossRef citations to date
0
Altmetric
Review

Developments in renal pharmacogenomics and applications in chronic kidney disease

, , &
Pages 251-266 | Published online: 28 Aug 2014

Abstract

Chronic kidney disease (CKD) has shown an increasing prevalence in the last century. CKD encompasses a poor prognosis related to a remarkable number of comorbidities, and many patients suffer from this disease progression. Once the factors linked with CKD evolution are distinguished, it will be possible to provide and enhance a more intensive treatment to high-risk patients. In this review, we focus on the emerging markers that might be predictive or related to CKD progression physiopathology as well as those related to a different pattern of response to treatment, such as inhibitors of the renin–angiotensin system (including angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers; the vitamin D receptor agonist; salt sensitivity hypertension; and progressive kidney-disease markers with identified genetic polymorphisms). Candidate-gene association studies and genome-wide association studies have analyzed the genetic basis for common renal diseases, including CKD and related factors such as diabetes and hypertension. This review will, in brief, consider genotype-based pharmacotherapy, risk prediction, drug target recognition, and personalized treatments, and will mainly focus on findings in CKD patients. An improved understanding will smooth the progress of switching from classical clinical medicine to gene-based medicine.

Natural history and epidemiology of chronic kidney disease (CKD)

CKD is defined as a reduced glomerular filtration rate (GFR), increased urinary albumin excretion, or both, and is a growing public health issue.Citation1 CKD progression is related to the GFR slope or key markers of renal damage (proteinuria or albuminuria) in diabetic patients. The Kidney Disease Outcome Quality Initiative defined CKD as the presence of renal impairment with a glomerular filtration rate (GFR) <60 mL/min.Citation2

The kidney is a key organ in the urinary system, acting as a filter of the blood, with homeostatic functions such as the regulation of electrolytes, control of blood pressure (BP), and maintenance of acid–base balance. It also modulates water imbalance, the reabsorption of several substances (such as glucose, water, and amino acids), and the excretion of urea and ammonium. Important hormones, calcitriol, erythropoietin, and the enzyme renin, are also synthesized by the kidney. In summary, kidney damage can contribute to disturbances in the equilibrium between exogenous and endogenous elements including drugs and metabolites.

CKD includes different types of renal disease. Glomerular disease is the main group, consisting of diabetic and hypertensive nephropathies, which are the leading causes of CKD in developed countries.Citation3 Other groups of CKD, such as glomerulonephritis and CKD of unknown causes (CKDU), are more common in countries in Asia and sub-Saharan Africa, and account for 10% of CKD worldwide and 16% in India.Citation4 Differences between countries are related mainly to chronic lifestyle-related diseases, decreased birth rates, and increased life expectancy in the developed regions. By contrast, infectious diseases continue to be prevalent in low-income countries, secondary to poor sanitation, inadequate supply of safe water, and high concentrations of disease-transmitting vectors.

There are many published studies focused on traditional risk-initiating factors such as ethnicity, sex, age, hyperfiltration, diabetes mellitus, familiar history of CKD, metabolic syndrome, albumin excretion, cardiovascular disease, primary kidney disease, and urological disorders. Progression factors such as BP, smoking, uric acid, nephrotoxins, anemia, hypertension, dyslipidemia, obesity, cardiovascular disease, proteinuria, inflammation, and hemostasis have also been evaluated.Citation5

Aside from the conventional factors, new markers have also been implicated in CKD pathogenesis and progression; these new markers include: adiponectin, kidney injury molecule-1, liver-type fatty acid binding protein, N-terminal pro-brain natriuretic peptide, factors involved in calcium–phosphate metabolism, A-type natriuretic peptide, adrenomedullin, neutrophil gelatinase-associated lipocalin, apolipoprotein A-IV, asymmetric dimethylarginine, and some recently identified genetic polymorphisms.Citation5

Prevalence of CKD is estimated to be 5%–16% worldwide.Citation6,Citation7 Complications include increased all-cause and cardiovascular mortality, kidney-disease progression, acute kidney injury, cognitive decline, anemia, mineral and bone disorders, and fractures.Citation8Citation10 The Epidemiology of Chronic Kidney Disease in Spain study estimated that approximately 10% of the Spanish adult population had some degree of CKD, and similar values were also estimated from other epidemiological studies.Citation11,Citation12 CKD is reasonably prevalent,Citation13 symptoms do not appear until they are at an advanced stage, and progress occurs over several years, leading to end-stage renal disease (esrD).Citation13 In this sense, early screening of CKD would be useful to facilitate diagnosis.

The incidence of esrD differs extensively worldwide: 400 per million population per year in Taiwan; 300 per million population per year in USA and Mexico, and 100–150 per million population per year in Europe.Citation14 Depending on the country, there are several markers and risk factors for the progression of CKD. The increasing prevalence of CKD generates concern about the cost of treatment for esrD. Bochud et alCitation15 describe that, despite the fact that prevalence of esrD is only about 0.2%, esrD programs now account for 6.7% of total medicare expenditure, and medicare costs associated with esrD increased by 57% between 1999 and 2004.

Considering the increasing prevalence and economic impact of CKD in previous decades, genetic studies have also been used to define the gene phenotypes involved in renal impairment, such as those related to high serum creatinine levels and GFR, hypertension, diabetic nephropathy, focal segmental glomerulosclerosis, albuminuria, and esrD.Citation17

Management issues in the treatment of CKD – clinical utility of pharmacogenomics

Here we review the link between CKD evolution and treatment, and we also identify the emerging markers and their pharmacogenomics.

Management in the treatment of CKD

The incidence of CKD, as a serious public health disease with a high morbidity and mortality, is increasing. Proteinuria is a predictor of outcome, but genetic factors have also been related to the progression of renal disease. CKD management remains a clinical challenge.

In fact, the renin–angiotensin–aldosterone system (RAAS) is a major pathway involved in the pathogenesis and progression of diabetic nephropathy,Citation18 and the blockade of RAAS, which improves urinary protein levels, has been proven to reduce the slope of GFR in nondiabetic experimental animals and humans compared with an intensified BP control.Citation19,Citation20

Classical treatments

We focused on two CKD preventive and therapeutic drugs and a complicating factor in CKD that are of potential interest in kidney pharmacogenomic applications: RAAS, Vitamin D receptor (VDR) agonists and salt-sensitivity.

RAAS inhibitors

Angiotensin converting enzyme (ACE) inhibitors and angiotensin II receptor blockers (ARBs) under similar conditions of blood pressure improve the progression of esrD and reduce the proteinuria rate better than non-RAAS antihypertensive drugs.Citation18,Citation21,Citation22 Captopril was the first ACE inhibitor that is effective in slowing the progression of diabetic nephropathy.Citation21 Indeed, RAAS inhibitors are considered for use in classical treatment for primary hypertension and involve two key approaches for CKD improvement, summarized by hemodynamic and antihypertensive changes as well as anti-inflammatory and antifibrotic properties. The main mechanism for both is the decrease of angiotensin II levels. With regard to inflammation, angiotensin II induces lymphocyte proliferation by nuclear factor-β (NF-kβ) activation.Citation23 Fibrosis is attenuated by induction of extracellular-matrix proteins via transforming growth factor-β.Citation24 On the other hand, hemodynamic beneficial effects are based on the maintenance of glomerular capillary hypertension by RAAS inhibitors.Citation25

VDR agonists

Several metabolic disturbances of CKD, such as acidosis, dyslipidemia, and vitamin D deficiency, could also be therapeutic targets for modifying the morbidity and mortality of CKD.Citation26

Patients with esrD show a deficiency of 1,25(OH)2D3 vitamin D and, consequently, often undergo vitamin D therapy.Citation27 In fact, this therapy has been beneficial in hemodialysis patients in terms of survival.Citation28 In this sense, the analogue of vitamin D has been shown to attenuate kidney interstitial fibrosisCitation29 and ameliorate glomerulosclerosis.Citation30 Analogues of vitamin D have also been related to a decrease in albuminuria or proteinuria in CKD.Citation31,Citation32

The beneficial effects of 1,25(OH)2D3 on BP and CKD progression are mediated by the NF-kβ pathwayCitation33 and by direct inhibition of 1,25(OH)2D3 on the RAAS.Citation34,Citation35 Both NF-kβ and RAAS are involved in immune response and related inflammation, oxidative stress, and fibrogenesis.Citation33,Citation36 Also, the prevention of secondary hyperparathyroidism by vitamin D treatment can ameliorate BP control.Citation37

Salt sensitivity

Salt sensitivity of BP is still not well defined. It is accepted that a person could be considered salt-sensitive when BP increases by 5%–10% after a large increase in dietary salt intake.Citation38 It has been described that dietary sodium intake have an impact on the efficacy of RAAS blockers in preventing CKD and cardiovascular disease.Citation39 On the other hand, RAAS blockers’ antiproteinuric effect is impaired in patients with high sodium intake. An observational trial described that increasing sodium intake was associated with a linear increase in the risk of progression of esrD.Citation40

Novel therapeutic approaches

Drugs focused on targeting inflammation and damaged systems (fibrosis, endothelin, oxidation, and advanced glycation end products) could be beneficial in preventing CKD progression.Citation41

Bardoxolone methyl and palmitoylethanolamide are new drugs for the treatment of CKD that target inflammation. Bardoxolone methyl, a first-in-class oral nuclear factor erythroid 2-related factor 2 agonist, seemed to have potential as a drug for improving renal function in advance diabetic nephropathy patients in a Phase II trial. However, in the Phase III study, the treatment had to be stopped due to emerging toxicity.Citation42,Citation43

On the other hand, palmitoylethanolamide belongs to a fatty acid ethanolamine family, and is a new and safe nonsteroidal, anti-inflammatory, and antifibrotic agent for CKDCitation44,Citation45 with activity at the peroxisome proliferator-activated receptor alpha.Citation46Citation48

Clinical utility of pharmacogenomics

There are numerous reasons to address the pharmacogenomics that are related to different kidney functions and treatments. Drug-treatment benefits in the pharmacogenomics of patients with kidney disease are based on avoiding nephrotoxic drugs, personalizing antihypertensive and cardiovascular drugs, and identifying the enzymes and proteins involved in the pharmacokinetics of drugs to improve renal function and BP.Citation15 Therefore, renal pharmacogenomics encompasses three important issues: ACE inhibitors, VDR agonists, and dietary salt intake.

Forty-four genes are included in the Pharmacogenomics Knowledge Database as very important pharmacogenes for their effects on renal function and diseases.Citation15 The most important genes involved in CKD disease are: CYP1A2 and CYP3A5; ABCB1; and methylenetetrahydrofolate reductase.

Phase I enzymes (CYP1A2 and CYP3A5)

The large interindividual variability in drug response is heritable,Citation49,Citation50 and single-nucleotide polymorphisms (SNPs) in genes encoding drug-metabolizing enzymes are involved in CKD.

CKDU constitutes 10% of CKD, and no specific causative agents have been identified. However, environmental toxins and heavy metals may be involved,Citation51,Citation52 such as persistent organic compounds that include polychlorinated biphenyls, organochlorine pesticides, and dioxins. Organic toxins are detoxified by cytochrome P450 enzymes being the CYP1A1, the enzyme that is most involved in the metabolism of persistent organic compounds. Several authors have reported the association of CYP1A1 polymorphism with various diseases such as diabetesCitation53 and neoplasia.Citation54Citation56 The prevalence of homozygous CYP1A1*2A mutants ranges between 2% and 18%, and for the heterozygous TC ranges between 32% and 55%. Siddarth et al performed a case-control study to evaluate the association of CYP1A1 in patients with CKD, and observed that subjects carrying at least one mutant allele of CYP1A1*2A (TC, CC) and *2C (AG, GG) had a 1.4- to twofold increased risk of CKDU as compared to those with the wild-type homozygous genotype, ie, TT (*2A) and AA (*2C).Citation57 However, other studies on Indian populations observed inter- and intra-ethnic variations of these two polymorphisms.Citation58Citation60

CYP1A2

CYP1A2 enzyme is responsible for 13% of the cytochrome P450 activity and has a large number of endogenous and exogenous substrates. There is a great amount of interindividual and inter-ethnic CYP1A2 variability due to both environmental and genetic factors. However, the mechanism of association between CYP1A2 and CKD is unknown, but probable mechanisms include an action mediated by CYP1A2 substrates.

Compared to other CYP-family genes, there is little data on CYP1A2 pharmacogenomics and antihypertensive drugs. The main attention has been focused on antipsychotic drugs, theophylline, and melatonin.Citation61 Antihypertensive-drug studies have been conducted with CYP2C9, which metabolizes different antihypertensive angiotensin II receptor antagonists, such as losartan, irbesartan, candesartan, and valsartan. CYP2C9 genotype has been shown to influence losartan metabolism, and response to irbesartan differed depending on CYP2C9 genotype.Citation62

Nutrigenomic studies have been performed to provide a mechanistic hypotheses for the relationship between CYP1A2 and BP. An increased risk of myocardial infarction with increased coffee consumption was reported among carriers of the CYP1A2 C variant.Citation63 Regular coffee or caffeine intake increases BP,Citation64 but there is a tolerance to the acute cardiovascular effects. There is no clear evidence that regular caffeine intake in the long-term increases the incidence of hypertension in the CYP1A2 carriers.Citation65,Citation66

CYP3A5

There are a large interindividual and inter-ethnic variations in CYP3A5*1 allele frequency. The CYP3A5 gene is associated with BP control, but further studies are needed to confirm the relationship with salt sensitivity in humans.Citation67 One hypothesized mechanism is the conversion of cortisol into 6 beta-hydroxycortisol, by CYP3A5, in the kidney. However, results are not conclusive. It would be of major interest to also clarify the putative role of CYP3A5 activity on intestinal drug disposition following various dietary salt intake levels.

CYP3A4 and CYP3A5 show similar substrate specificity for each of amlodipine, felodipine, nicardipine, nifedipine, atorvastatin, pravastatin, cerivastatin, lovastatin, celiprolol, digoxin, diltiazem, enalapril, losartan, and verapamil.Citation68,Citation69 However, the majority of pharmacogenetic studies are concentrated on tacrolimus and cyclosporine as CYP3A5 genotypes clearly influence the pharmacokinetics of the immunosuppressant tacrolimus.Citation70 Only a few studies with small sample sizes have analyzed the role of CYP3A5 variants on the response to drugs used to treat cardiovascular conditions. CYP3A5 variants appear to influence the pharmacokinetics of statins,Citation71 and CYP3A5*1 carriers may experience a diminished pharmacological effect of verapamil.Citation72 Eap et al studied the combined action of CYP3A5 and ABCB1 variants on BP, and observed that there was an association with altered response to lisinopril.Citation73 A study of plasma amlodipine concentrations in 40 healthy Korean men observed that carriers of the CYP3A5*3/*3 genotype had lower levels of amlodipine than CYP3A5*1 carriers, but the BP decrease was similar in both groups.Citation74

Transporters (ABCB1)

The ABCB1 gene encodes the P-glycoprotein (also named as Pgp, MDR1, and ABCB1), which belongs to the superfamily of human ABC transporters. It is also known as the multidrug resistance gene, and several ABCB1 genetic variants have been shown to influence Pgp expression in humans, including the 3435 C>T and 2677 G>T variants. Pgp is an efflux pump that transports endogenous substrates (eg, steroids, lipids, phospholipids, and cytokines), drugs (eg, digoxin, cyclosporine, tacrolimus, diltiazem, verapamil, etc), and other exogenous substrates out of the cells.Citation75 ABCB1 polymorphisms have been widely studied in transplant patients treated with cyclosporine, and it was observed that TT carrier patients on C3435T, G2677T, and C1236T SNPs (Pgp-low pumpers) showed lower Pgp activity than noncarriers.Citation76

SNPs related to drug transporters have also been described in CKD patients. The C3435T SNP in the gene of ABCB1 that codify P-glycoprotein was correlated with renal function and BP in two Chinese populations.Citation77 Patients with TT genotype showed an increased risk of CKD, and higher systolic BP and pulse pressure. Results were similar in elderly subjects, with CKD with a higher risk of CKD progression and hypertension. These authors concluded the importance of ABCB1 SNP in CKD specially in elderly population. The regulation of Pgp expression seems to be influenced by multiple nuclear receptors: namely, constitutive androstane receptor-betaCitation78 and VDR.Citation79Citation81

Although the role of ABCB1 genes are widely known in the field of transplant patients, the application in CKD progression and BP regulation is still not well defined.

Nuclear receptors (VDR and PXR)

VDR is widely expressed in the human kidney, namely in the epithelial cells of the proximal and distal tubules, collecting duct, and glomerulus.Citation82 VDR is a ligand-induced nuclear receptor that regulates the expression of over 900 genes throughout the genome,Citation83,Citation84 such as ABCB1,Citation79Citation81 CYP24A1,Citation84 CYP3A4,Citation85 CYP3A7, FGF23,Citation86 and SLC34A3. Most studies have attempted to correlate VDR polymorphisms with the development of secondary hyperparathyroidism.Citation87 Grzegorzewska and Ostromecki described the distribution of variants of vitamin D-binding protein gene, VDR with respect to PTH serum concentrations, and response to cinacalcet treatment in patients with secondary hyperparathyroidism.Citation88 Other studies have investigated the association of polymorphisms in the VDR gene with protection against esrD and periodontitis.Citation89

Variants of VDR and variants within the VDR gene may influence renal function and BP, but there is a lack of conclusive data on the association with renal function. In a study of people of Indian and African descent, vitamin D deficiency was significantly associated with increased diastolic BP and triglyceride levels, and reduced high-density lipoprotein cholesterol.Citation90 There is evidence of associations between VDR variants and diabetes, which is a major CKD risk factor. Randomized controlled trials have provided convincing evidence that VDR agonists confer renoprotection in humans.Citation91Citation94

Although PXR is not currently considered to be a gene associated with BP or renal function, its role in controlling the expression of genes such as ABCB1 and CYP3A5, its involvement in steroid hormone metabolism, its action on lipid and energy metabolism, its action on inflammation, as well as its interaction with VDR all point toward PXR being an important player in kidney diseases. The role of PXR as a xenobiotic and endobiotic sensor, its ability to bind to a large array of ligands, and its numerous transcriptional gene targets suggest that PXR may mediate complex gene– environment, drug–environment and drug–drug interactions with important consequences on human health, including kidney function.

ACE gene

The ACE gene encodes ACE, an enzyme involved in the RAAS and which plays a key role in BP control. There is a high interindividual variability in circulating ACE levels, with the 287-bp Alu-repeat sequence insertion/deletion polymorphism located in intron 16 (ACE I/D) being the most extensively studied ACE genetic variant, with more than 4,000 publications during the past 20 years.Citation95 Other ACE variants have been described, and the genetic diversity of ACE is particularly high in people of African descent.Citation96Citation98 The associations of the ACE I/D polymorphism with hypertension and cardiovascular disease have been inconsistent. Some authors suggest that testing for the ACE I/D polymorphism is useful for predicting the renoprotective effect of ACE inhibitor or angiotensin-receptor blocker treatment in patients with kidney disease.Citation99 There is currently no evidence to support a role of the ACE I/D polymorphism in predicting future risk of cardiovascular events or BP response to ACE inhibitors in the absence of renal dysfunction.

One of the first pharmacogenetic studies evaluating efficacy variability of ACE inhibitors on albumin excretion rates in nonhypertensive insulin-dependent patients with normoalbuminuria or microalbuminuria was conducted in 1998.Citation101 The application of ACE polymorphisms has been confirmed by different authors.Citation100 Patients carrying the II genotype had a higher albuminuria reduction and better BP control.Citation101,Citation102 In type I diabetic patients, the I allele had the best outcomes in terms of renal phenotypes (with decline in albuminuria and decreased BP),Citation103Citation105 and type 2 diabetic patients with the II genotype plus ID alleles had decreased mortality, esrD, and diabetes progression.Citation106 However, conflicting data exist as some other authors did not find a correlation between BP and ACE variants.Citation107Citation109 ARB-treatment outcomes have also been evaluated in different studies, and the data shows that DD genotype carriers have diminished renoprotection.Citation105,Citation110 A recent metaregression analysis evaluated 129 papers to study the effect of ACE I/D polymorphisms on CKD risk and concluded that the D allele had the highest risk for CKD in hypertensive Asian males (odds ratio, 3.75).

Review of pathogenesis: specific genetic polymorphisms in CKD

Pathogenesis

CKD is a complex pathophysiologic process resulting from multiple etiologies. It is classified as a multifactorial disease secondary to a combination of genetic and environmental factors that influence the onset and development of esrD.Citation111 Risk factors for development of CKD could be gathered in two groups: susceptibility to kidney disease due to sociodemographic and genetic factors; and exposure to variables that can initiate kidney disease.

The main risk factors for CKD described in the literature include hypertension, obesity, and diabetes. The global prevalence of hypertension in adults was estimated to be about 26% (972 million cases) in 2000.Citation112 Prevalence of hypertension is higher in urban populations than in rural populations in developing countries (global prevalence is 639 million [66%]).Citation113 The worldwide prevalence, adjusted for age and sex, is projected to increase to 1.56 billion by 2025. Therefore, treatment of hypertension is one of the most important interventions in the pharmacological management of CKD. Similar trends are apparent for diabetes and obesity.Citation114,Citation115 The worldwide prevalence of diabetes in adults is estimated to be 6.4%, affecting 285 million people, and is expected to rise to 7.7% by 2030 (439 million cases). The increase in overweight and obese children is particularly alarming.Citation114 Obesity raises BP physical compression of the kidneys by increasing renal tubular sodium reabsorption, impairing pressure natriuresis, and by activating the sympathetic nervous system and RAAS.Citation116

Genetic susceptibility is an important determining factor for the onset and/or progression of esrD and its complications, and different studies have identified new susceptibility loci for reduced renal function.Citation117 Although environmental risk factors and interactions between genes and environment undoubtedly play an additional role, nontraditional risk factors such as oxidative stress, inflammation, and immune processes may be important contributors to the pathogenesis and progression to esrD.

Polymorphisms in CKD pathogenesis

Here we present genetic polymorphisms related to CKD and the progression to esrD ().

Table 1 Characteristics and results of the main published studies involving gene polymorphisms and effect on renal function

Inflammation could be a causal factor in the development of CKD and may be established before the onset of renal disease. The inflammatory response involved in renal damage produces proinflammatory cytokines and chemokines, an increase of leukocytes, intensification of interstitial nephritis, and a progression of fibrosis. Recent studies have suggested roles for toll-like receptor 9 (TLR-9) in the development of renal diseases such as glomerulonephritis,Citation118 lupus nephritis,Citation119 and the progression of immunoglobulin A nephropathy, and have also suggested that TLR-9 could be associated with severe clinical phenotypes.Citation120,Citation121 A case-control study observed significantly different allelic distribution of 1237T/C, but not 1486T/C or 1635G/A, between esrD patients and controls. Higher GFR values for patients with the TLR-9-1237TT genotype were obtained, but differences were not statistically significant.Citation122

Associations of 48 chemokine gene variants with esrD have been tested; however, the small sample size did not allow to detect moderate effects. The authors found association between esrD and four SNPs.Citation123 Two of them expressed protection (interleukin 4 receptor A/G and CCL2) and the other two expressed susceptibility (STAT4 binding site and nitric oxide synthase 3). However, after adjusting for multiple testing, the results were not significant. Singh et al found a significant association between a high risk for esrD and both CXCL2G801A and CXCR2, whereas CCL2I/D showed a reduced risk for esrD.Citation124 In the process of CKD progression to the terminal stage, the cytokine-mediating angiotensin action (transforming growth factor-β1) could be also related as it is involved in the process of tissue sclerosis. With regard to that, Nabrdalik et al performed a case-control study and identified the mutant C allele as the related allele with the higher risk of CKD (twofold elevated risk).Citation125 These results were similar to those observed by Buraczynska et al and Coll et al.Citation126,Citation127 In contrast, other authors did not find an association between transforming growth factor-β1 and CKD occurrence.Citation128Citation130

Hypertension is second to diabetes as the leading independent cause of esrD. There is available evidence supporting the association of genetic variants of the RAAS and pharmacogenetic responses. ACE gene polymorphism (ACE-ID) has been associated with higher circulating plasma ACE concentrations,Citation131 and the molecular variant M235T of the AGT gene has been associated with higher plasma AGT levels in patients homozygous for the T allele.Citation132 Polymorphisms related to treatment efficacy with ACE inhibitors and angiotensin II receptor blockers (ARBs) are explained in . Several studies have linked variants of AGTR1 with hypertension and heart diseases, but conclusive data related to esrD is lacking. The reduction of BP by ACE inhibitors is mediated by decreased formation of the vasoconstrictor angiotensin II, and by increased levels of the vasodilator bradykinin and endothelial nitric oxide synthase (eNOS). Bradykinin receptor B2 and eNos gene polymorphism could affect the response to ACE inhibitors, and was studied by Silva et al.Citation133 The results showed that the C allele for eNOS and TT genotype for the bradykinin receptor B2 were more frequent in good responders.

The MYH9 gene encodes for the non-muscle myosin IIA heavy chain, a subunit of myosin IIA protein that is involved in several functions including cytokinesis, cell motility, maintenance of cell shape, and secretion. Recent studies have linked MYH9 SNPs/haplotypes to a risk of developing focal segmental glomerulosclerosis, hypertensive nephropathy, and nondiabetic esrD among African and Hispanic Americans.Citation134Citation138 Because African Americans seem to have greater risk than European Americans, even after considering socioeconomic status, a genome-wide admixture scan was performed to find genetic risk alleles. MYH9 genes were associated with two- to four-times greater risk of nondiabetic esrD. Multiple clusters of SNPs was performed and remained significant.Citation137 As African Americans showed a higher risk for specific forms of CKD, the influence of MYH9 SNPs was studied in African ancestry populations. In diabetic participants, the G allele of rs5756152 was associated with a significant decrease in serum creatinine and GFR, but not in nondiabetic patients. Interactions by diabetic status were also significant, but rs12107 analysis showed no association with renal function.Citation140

In a Spanish cohort of healthy elderly individuals, Tavira et al studied the effect of common MYH9 SNPs on renal function. The multivariate analysis showed that age, diabetes, sex, and the MYH9 genotype were risk factors for GFR <60 mL/min/1.73 m2. MYH9 rs3752462 T carriers had lower mean GFR compared to CC homozygotes.Citation141 Aside from MYH9 polymorphisms, variations in the gene encoding apolipoprotein L1 (APOL1) have recently been shown to be associated with kidney disease in African Americans.Citation142Citation145 Fifteen MYH9 SPNs and two APOL1 SNPs were studied in association with type 2 diabetes mellitus in European Americans. As reported previously, MYH9 polymorphisms were associated with type 2 diabetes mellitus esrD susceptibility, and APOL1 could not be tested for as the frequency was not appreciable.Citation139 APOL1 variants have previously been studied and it was concluded that these polymorphisms were not present in European Americans or in Europeans.Citation146 Polymorphisms in APOL1 related with CKD progression has been evaluated in two populations: black African Americans with kidney disease and hypertension (African American Study of Kidney Disease and Hypertension); and black and white African and European Americans with chronic renal insufficiency (Chronic Renal Insufficiency Cohort study). Black subjects in the APOL1 high-risk group had a more rapid decline in GFR and a higher risk of composite renal outcome, defined as doubling serum creatinine or incident of esrD, compared to white patients.

Developments in pharmacogenomics – clinical implications and personalized therapies

In the last few decades, despite the fact that CKD diagnosis and progression risks are still based on clinical observations, there have been notable advances in genetic studies that have led to a more selective set of therapeutic strategies.Citation147 Herein, we review some of the potential applications of genotyping that have benefits for CKD risk prediction, guided treatment, and guided individualized strategies.

Nowadays, diagnosis and determination of risk factors based on genotyping is available for many disorders such as breast cancer (by testing BRCA 1 and 2).Citation150 However, in CKD patients, definitive DNA-based diagnostics and risk prediction have not yet been made available.Citation17

However, recent studies in African-American patients of nondiabetic CKD have profiled possible risk factors in polymorphisms of apolipoprotein L-I genesCitation136,Citation144,Citation145 and of the non-muscle myosin heavy chain type II isoform A.Citation136,Citation137

The benefits derived from pharmacogenetics are aimed at individualizing treatments, reducing drug side effects, and improving drug efficacy. For example, the effects of polymorphism of genes in the cytochrome P450 enzyme complex are widely known, and genetic tests for this polymorphism before receiving treatments are routinely practiced.Citation151,Citation152 Also, patients with stage 5 esrD undergoing hemodialysis may present allelic variants in the cytochrome P450 2D6 (CYP2D6), which would be determinant in the metabolism of codeine into morphine, thus pointing to different susceptibility to morphine.Citation153 Other therapeutic applications focusing on treatment with atenolol and enalapril have been identified involving polymorphism of ACE; the suggestion of these applications is that homozygous patients with a deletion allele may benefit from other antihypertensive drugs.Citation154 These novel applications in pharmacogenomics that involve personalized treatment of patients with CKD provide clinicians with key management tools that are far from classical treatments.

Studying genetic variants may be helpful for personalized therapeutic strategies. Those genetic variants that would allow prediction of risks relative to drug exposure could guide physicians in evaluating cost-effectiveness of new strategies in CKD patients. For example, deletion polymorphism in the CC-chemokine receptor 5 (CCR5) gene (CCR5Δ32), which is known to cause a dysfunctional CCR5 protein, has been related to an improvement of inflammatory state with better survival.Citation155,Citation156 Therefore, patients with esrD undergoing hemodialysis and, thus, suffering from a well-known persistent inflammatory state (ie, wasting syndrome) would potentially benefit from a blockade of CCR5 based therapy.Citation18,Citation157 Glutathione S-transferase M1 (GST M1) null allele is another polymorphism related to CKD progression caused by deletion in the GST M1 gene and related to oxidative damage protection.Citation158,Citation159 In fact, hemodialysis patients are exposed to a higher oxidative stress status, and it has been shown that hemodialysis patients homozygous for the GST M1 null allele had higher risk for death compared with those who possess GST M1 activity.Citation160

Interestingly, uremic milieu (which includes oxidative stress), wasting syndrome, inflammation, anemia, vascular calcification, and concerns associated with dialysis would render patients with CKD more susceptible to genetic variants.Citation155,Citation161Citation163 Knowing these interactions of the gene environment could be helpful in clinical practice in addressing patients’ nutritional habits and lifestyle.Citation164 In fact, renal-function impairment per se has been considered an environment factor, varying the effect of two polymorphisms of RAS (AGTR1 A1166C and ACE insertion/deletion) on left ventricular hypertrophy.Citation165Citation170 Another example of gene– environment interaction that has been established is in the susceptibility of some patients to respond to ACE inhibitors depending on dietary sodium intake and the ACE deletion/deletion genotype, which would encourage the prescription of a salt-restricted diet in those patients with this genotype variant.Citation171

It has been questioned whether epigenetics is involved in esrD progression.Citation172 In fact, inflammation, atherosclerotic processes, and aging are related to DNA methylation and act as catalyzers in the poor prognosis of the cardiovascular disease in dialysis patients.Citation155,Citation161,Citation172,Citation173 These findings highlight the importance of targeting the epigenome with epigenetic drugs. Therapeutic applications of epigenetics in patients with CKD, and even in patients with esrD, could be focused on RNA interference, which is crucial in renal homeostasis, mainly linked to podocyte dedifferentiation-related proteinuria.Citation174Citation177 Hyperhomocysteinemia has been described in CKD and esrD patients.Citation173,Citation178 These patients showed high levels of S-adenosylhomocysteine, causing hypomethylation of DNA.

Thus, glomerular and interstitial fibrosis could be related to epigenetic modifications trough transcriptional regulation.Citation77,Citation179Citation181 The interest in epigenetics in clarifying hypertensive and diabetic CKD is increasing.

In this sense, as CKD incidence has been increasing in recent years, utilizing the potential therapeutic benefits of interventions to prevent esrD may be helpful in reducing the economic and clinical impact of CKD.Citation18

Conclusions

Interesting approaches to in-depth pharmacogenetics are increasing for tailored medicine. In fact, nowadays, high-risk CKD patients could be detected by genotype information. Nevertheless, these techniques assume high cost and must be optimized alongside classical clinical tests. Advances in technology in gene sequencing with epigenetic investigation, as well as well-designed studies on gene–gene interactions, gene–environment interactions, and DNA modifications (epigenetics), may improve our knowledge of CKD-related genes and subsequent patient care. These genetic markers could be useful for the prediction of CKD progression, but clinical risk factors remain more valuable in terms of prediction. However, further technical developments and epidemiological experimental data are still needed to demonstrate and establish the most cost-effective approach. The bulk of information in pharmacogenomics published nowadays is targeted at expanding this field of information, although physicians must be critical in their analysis and interpretation of the results.

Given that CKD is a complex disorder, and that it can benefit from genetic testing, further challenges include studying proteins, transcripts, and metabolites in order to correlate them with genetic data, improve clinical outcome, and lead to routine genetic tests in the clinical care of CKD.

Acknowledgments

Núria Lloberas is a researcher in ISCIII Miguel Servet (CP06/00067) and REDinREN RD12/0021/003.

Disclosure

The authors report no conflicts of interest in this work.

References

  • LeveyASAtkinsRCoreshJChronic kidney disease as a global public health problem: approaches and initiatives – a position statement from Kidney Disease Improving Global OutcomesKidney Int200772324725917568785
  • TaalMWChronic Kidney Disease in General Populations and Primary CareCurr Opin Nephrol Hypertens201322659359824104481
  • JhaVGarcia-GarciaGIsekiKChronic kidney disease: global dimension and perspectivesLancet2013382988826027223727169
  • BallSLloydJCairnsTWhy is there so much end-stage renal failure of undetermined cause in UK Indo-Asians?QJM200194418719311294961
  • KronenbergFEmerging risk factors and markers of chronic kidney disease progressionNat Rev Nephrol200951267768919935815
  • HallanSICoreshJAstorBCInternational comparison of the relationship of chronic kidney disease prevalence and ESRD riskJ Am Soc Nephrol20061782275228416790511
  • ChadbanSJBrigantiEMKerrPGPrevalence of kidney damage in Australian adults: The AusDiab kidney studyJ Am Soc Nephrol2003147 Suppl 2S131S13812819318
  • BowlingCBInkerLAGutiérrezOMAge-specific associations of reduced estimated glomerular filtration rate with concurrent chronic kidney disease complicationsClin J Am Soc Nephrol20116122822282822034504
  • DalrympleLSMohammedSMMuYRisk of cardiovascular events after infection-related hospitalizations in older patients on dialysisClin J Am Soc Nephrol2011671708171321566109
  • GoASChertowGMFanDMcCullochCEHsuCYChronic kidney disease and the risks of death, cardiovascular events, and hospitalizationN Engl J Med2004351131296130515385656
  • CoreshJSelvinEStevensLAPrevalence of chronic kidney disease in the United StatesJAMA2007298172038204717986697
  • OteroAde FranciscoAGayosoPGarcíaFEPIRCE Study GroupPrevalence of chronic renal disease in Spain: results of the EPIRCE studyNefrologia2010301788620038967
  • TaalMWScreening for chronic kidney disease: preventing harm or harming the healthy?PLoS Med2012911e100134523185137
  • HallanSIVikseBERelationship between chronic kidney disease prevalence and end-stage renal disease riskCurr Opin Nephrol Hypertens200817328629118408480
  • BochudMBurnierMGuessousITop Three Pharmacogenomics and Personalized Medicine Applications at the Nexus of Renal Pathophysiology and Cardiovascular MedicineCurr Pharmacogenomics Person Med20119429932223049672
  • FoleyRNCollinsAJEnd-stage renal disease in the United States: an update from the United States Renal Data SystemJ Am Soc Nephrol200718102644264817656472
  • DrawzPESedorJRThe genetics of common kidney disease: a pathway toward clinical relevanceNat Rev Nephrol20117845846821712849
  • RuggenentiPCravediPRemuzziGThe RAAS in the pathogenesis and treatment of diabetic nephropathyNat Rev Nephrol20106631933020440277
  • RossingPHommelESmidtUMParvingHHReduction in albuminuria predicts diminished progression in diabetic nephropathyKidney Int Suppl199445S145S1498158884
  • ApperlooAJde ZeeuwDde JongPEShort-term antiproteinuric response to antihypertensive treatment predicts long-term GFR decline in patients with non-diabetic renal diseaseKidney Int Suppl199445S174S1788158890
  • LewisEJHunsickerLGBainRPRohdeRDThe effect of angiotensin-converting-enzyme inhibition on diabetic nephropathy. The Collaborative Study GroupN Engl J Med199332920145614628413456
  • BrennerBMCooperMEde ZeeuwDEffects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathyN Engl J Med20013451286186911565518
  • SowersJRWhaley-ConnellAEpsteinMNarrative review: the emerging clinical implications of the role of aldosterone in the metabolic syndrome and resistant hypertensionAnn Intern Med20091501177678319487712
  • RusterCWolfGRenin-angiotensin-aldosterone system and progression of renal diseaseJ Am Soc Nephrol200617112985299117035613
  • TaalMWBrennerBMRenoprotective benefits of RAS inhibition: from ACEI to angiotensin II antagonistsKidney Int20005751803181710792600
  • LiuSVaziriNDRole of PCSK9 and IDOL in the pathogenesis of acquired LDL receptor deficiency and hypercholesterolemia in nephrotic syndromeNephrol Dial Transplant201429353854324166456
  • LevinABakrisGLMolitchMPrevalence of abnormal serum vitamin D, PTH, calcium, and phosphorus in patients with chronic kidney disease: results of the study to evaluate early kidney diseaseKidney Int2007711313817091124
  • WolfMBetancourtJChangYImpact of activated vitamin D and race on survival among hemodialysis patientsJ Am Soc Nephrol20081971379138818400938
  • TanXLiYLiuYParicalcitol attenuates renal interstitial fibrosis in obstructive nephropathyJ Am Soc Nephrol200617123382339317082242
  • MakibayashiKTatematsuMHirataMA vitamin D analog ameliorates glomerular injury on rat glomerulonephritisAm J Pathol200115851733174111337371
  • FishbaneSChittineniHPackmanMDutkaPAliNDurieNOral paricalcitol in the treatment of patients with CKD and proteinuria: a randomized trialAm J Kidney Dis200954464765219596163
  • SzetoCCChowKMKwanBCChungKYLeungCBLiPKOral calcitriol for the treatment of persistent proteinuria in immunoglobulin A nephropathy: an uncontrolled trialAm J Kidney Dis200851572473118436082
  • BonizziGKarinMThe two NF-kappaB activation pathways and their role in innate and adaptive immunityTrends Immunol200425628028815145317
  • ResnickLMMüllerFBLaraghJHCalcium-regulating hormones in essential hypertension. Relation to plasma renin activity and sodium metabolismAnn Intern Med198610556496543532893
  • LiYCKongJWeiMChenZFLiuSQCaoLP1,25-Dihydroxyvitamin D(3) is a negative endocrine regulator of the renin-angiotensin systemJ Clin Invest2002110222923812122115
  • GuijarroCEgidoJTranscription factor-kappa B (NF-kappa B) and renal diseaseKidney Int200159241542411168923
  • RichartTLiYStaessenJARenal versus extrarenal activation of vitamin D in relation to atherosclerosis, arterial stiffening, and hypertensionAm J Hypertens20072091007101517765144
  • SanadaHJonesJEJosePAGenetics of salt-sensitive hypertensionCurr Hypertens Rep2011131556621058046
  • Lambers HeerspinkHJHoltkampFAParvingHHModeration of dietary sodium potentiates the renal and cardiovascular protective effects of angiotensin receptor blockersKidney Int201282333033722437412
  • VegterSPernaAPostmaMJNavisGRemuzziGRuggenentiPSodium intake, ACE inhibition, and progression to ESRDJ Am Soc Nephrol201223116517322135311
  • TurnerJMBauerCAbramowitzMKMelamedMLHostetterTHTreatment of chronic kidney diseaseKidney Int201281435136222166846
  • PergolaPEKrauthMHuffJWEffect of bardoxolone methyl on kidney function in patients with T2D and Stage 3b-4 CKDAm J Nephrol201133546947621508635
  • PergolaPERaskinPTotoRDBardoxolone methyl and kidney function in CKD with type 2 diabetesN Engl J Med2011365432733621699484
  • IndraccoloUBarbieriFEffect of palmitoylethanolamide-polydatin combination on chronic pelvic pain associated with endometriosis: preliminary observationsEur J Obstet Gynecol Reprod Biol20101501767920176435
  • TruiniABiasiottaADi StefanoGPalmitoylethanolamide restores myelinated-fibre function in patients with chemotherapy-induced painful neuropathyCNS Neurol Disord Drug Targets201110891692022229320
  • YoshiharaDKugitaMYamaguchiTGlobal Gene Expression Profiling in PPAR-γ Agonist-Treated Kidneys in an Orthologous Rat Model of Human Autosomal Recessive Polycystic Kidney DiseasePPAR Res2012201269589822666229
  • ParkCWZhangYZhangXPPARalpha agonist fenofibrate improves diabetic nephropathy in db/db miceKidney Int20066991511151716672921
  • MerkDSchubert-ZsilaveczMNew hope or drawbacks: will chronic kidney disease be treatable with small molecules in the near future?Future Med Chem20124326927122393934
  • WangLMcLeodHLWeinshilboumRMGenomics and drug responseN Engl J Med2011364121144115321428770
  • RahmiogluNHeatonJClementGGenetic epidemiology of induced CYP3A4 activityPharmacogenet Genomics2011211064265121750469
  • SoderlandPLovekarSWeinerDEBrooksDRKaufmanJSChronic kidney disease associated with environmental toxins and exposuresAdv Chronic Kidney Dis201017325426420439094
  • RajapurkarMMJohnGTKirpalaniALWhat do we know about chronic kidney disease in India: first report of the Indian CKD registryBMC Nephrol2012131022390203
  • WangXLGrecoMSimASDuarteNWangJWilckenDEEffect of CYP1A1 MspI polymorphism on cigarette smoking related coronary artery disease and diabetesAtherosclerosis2002162239139711996959
  • SergentanisTNEconomopoulosKPFour polymorphisms in cytochrome P450 1A1 (CYP1A1) gene and breast cancer risk: a meta-analysisBreast Cancer Res Treat2010122245946920035380
  • WrightCMLarsenJEColosimoMLGenetic association study of CYP1A1 polymorphisms identifies risk haplotypes in nonsmall cell lung cancerEur Respir J201035115215919608585
  • ChenJChengMYiLJiangCBRelationship between CYP1A1 genetic polymorphisms and renal cancer in ChinaAsian Pac J Cancer Prev20111292163216622296350
  • SiddarthMDattaSKAhmedRSBanerjeeBDKalraOPTripathiAKAssociation of CYP1A1 gene polymorphism with chronic kidney disease: a case control studyEnviron Toxicol Pharmacol201336116417023619522
  • SobtiRCSharmaSJoshiAJindalSKJanmejaAGenetic polymorphism of the CYP1A1, CYP2E1, GSTM1 and GSTT1 genes and lung cancer susceptibility in a North Indian populationMol Cell Biochem20042661–21915646021
  • SreejaLSyamalaVHariharanSMadhavanJDevanSCAnkathilRPossible risk modification by CYP1A1, GSTM1 and GSTT1 gene polymorphism in lung cancer susceptibility in a south Indian populationJ Hum Genet2005501261862716228113
  • AnantharamanDChaubalPMKannanSBhiseyRAMahimkarMBSusceptibility to oral cancer by genetic polymorphisms at CYP1A1, GSTM1 and GSTT1 loci among Indians: tobacco exposure a risk modulatorCacinogenesis200728714551462
  • GunesADahlMLVariation in CYP1A2 activity and its clinical implications: influence of environmental factors and genetic polymorphismsPharmacogenomics20089562563718466106
  • HallbergPKarlssonJKurlandLThe CYP2C9 genotype predicts the blood pressure response to irbesartan: results from the Swedish Irbesartan Left Ventricular Hypertrophy Investigation vs Atenolol (SILVHIA) trialJ Hypertens200220102089209312359989
  • CornelisMCMondaKLYuKGenome-wide meta-analysis identifies regions on 7p21 (AHR) and 15q24 (CYP1A2) as determinants of habitual caffeine consumptionPLoS Genet201174e100203321490707
  • NoordzijMUiterwaalCSArendsLRKokFJGrobbeeDEGeleijnseJMBlood pressure response to chronic intake of coffee and caffeine: a meta-analysis of randomized controlled trialsJ Hypertens200523592192815834273
  • KlagMJWangNYMeoniLACoffee intake and risk of hypertension: the Johns Hopkins precursors studyArch Intern Med2002162665766211911719
  • WinkelmayerWCStampferMJWillettWCCurhanGCHabitual caffeine intake and the risk of hypertension in womenJAMA2005294182330233516278361
  • BochudMBovetPBurnierMEapCBCYP3A5 and ABCB1 genes and hypertensionPharmacogenomics200910347748719290795
  • WilkinsonGRDrug metabolism and variability among patients in drug responseN Engl J Med2005352212211222115917386
  • SiestGJeannessonEVisvikis-SiestSEnzymes and pharmacogenetics of cardiovascular drugsClin Chim Acta20073811263117362901
  • DalyAKSignificance of the minor cytochrome P450 3A isoformsClin Pharmacokinet2006451133116430309
  • WillrichMAHirataMHHirataRDStatin regulation of CYP3A4 and CYP3A5 expressionPharmacogenomics20091061017102419530969
  • JinYWangYHMiaoJCytochrome P450 3A5 genotype is associated with verapamil response in healthy subjectsClin Pharmacol Ther200782557958517443134
  • EapCBBochudMElstonRCCYP3A5 and ABCB1 genes influence blood pressure and response to treatment, and their effect is modified by saltHypertension20074951007101417372036
  • KimKAParkPWLeeOJEffect of CYP3A5*3 genotype on the pharmacokinetics and pharmacodynamics of amlodipine in healthy Korean subjectsClin Pharmacol Ther200680664665617178265
  • KimRBDrugs as P-glycoprotein substrates, inhibitors, and inducersDrug Metab Rev2002341–2475411996011
  • LlaudóIColomHGiménez-BonaféPDo drug transporter (ABCB1) SNPs and P-glycoprotein function influence cyclosporine and macrolides exposure in renal transplant patients? Results of the pharmacogenomic substudy within the symphony studyTranspl Int201326217718623216707
  • LiuMLiYCitterioLA functional common polymorphism of the ABCB1 gene is associated with chronic kidney disease and hypertension in ChineseAm J Hypertens201326121428143623926124
  • BurkOArnoldKAGeickATegudeHEichelbaumMA role for constitutive androstane receptor in the regulation of human intestinal MDR1 expressionBiol Chem2005386650351316006237
  • TachibanaSYoshinariKChikadaTToriyabeTNagataKYamazoeYInvolvement of Vitamin D receptor in the intestinal induction of human ABCB1Drug Metab Dispos20093781604161019460946
  • SaekiMKuroseKTohkinMHasegawaRIdentification of the functional vitamin D response elements in the human MDR1 geneBiochem Pharmacol200876453154218602086
  • ChowECDurkMRCumminsCLPangKS1Alpha,25-dihydroxyvitamin D3 up-regulates P-glycoprotein via the vitamin D receptor and not farnesoid X receptor in both fxr(−/−) and fxr(+/+) mice and increased renal and brain efflux of digoxin in mice in vivoJ Pharmacol Exp Ther2011337384685921421739
  • KumarRSchaeferJGrandeJPRochePCImmunolocalization of calcitriol receptor, 24-hydroxylase cytochrome P-450, and calbindin D28k in human kidneyAm J Physiol19942663 Pt 2F477F4858160797
  • SchusterICytochromes P450 are essential players in the vitamin D signaling systemBiochim Biophys Acta20111814118619920619365
  • WangTTTavera-MendozaLELaperriereDLarge-scale in silico and microarray-based identification of direct 1,25-dihydroxyvitamin D3 target genesMol Endocrinol200519112685269516002434
  • MakishimaMLuTTXieWVitamin D receptor as an intestinal bile acid sensorScience200229655711313131612016314
  • BarthelTKMathernDRWhitfieldGK1,25-Dihydroxyvitamin D3/VDR-mediated induction of FGF23 as well as transcriptional control of other bone anabolic and catabolic genes that orchestrate the regulation of phosphate and calcium mineral metabolismJ Steroid Biochem Mol Biol20071033–538138817293108
  • SantoroDCaccamoDGagliostroGVitamin D metabolism and activity as well as genetic variants of the vitamin D receptor (VDR) in chronic kidney disease patientsJ Nephrol201326463664422976524
  • GrzegorzewskaAEOstromeckiGGene polymorphism of the vitamin D receptor, vitamin D-binding protein and calcium-sensing receptor in respect of calcium-phosphate disturbances in chronic dialysis patientsPrzegl Lek2013709735738 Polish24455835
  • de SouzaCMBraosiAPLuczyszynSMAssociation between vitamin D receptor gene polymorphisms and susceptibility to chronic kidney disease and periodontitisBlood Purif2007255–641141917914260
  • Vélayoudom-CéphiseFLLariflaLDonnetJPVitamin D deficiency, vitamin D receptor gene polymorphisms and cardiovascular risk factors in Caribbean patients with type 2 diabetesDiabetes Metab201137654054521764620
  • AlborziPPatelNAPetersonCParicalcitol reduces albuminuria and inflammation in chronic kidney disease: a randomized double-blind pilot trialHypertension200852224925518606901
  • AgarwalRAcharyaMTianJAntiproteinuric effect of oral paricalcitol in chronic kidney diseaseKidney Int20056862823282816316359
  • de ZeeuwDAgarwalRAmdahlMSelective vitamin D receptor activation with paricalcitol for reduction of albuminuria in patients with type 2 diabetes (VITAL study): a randomised controlled trialLancet201037697521543155121055801
  • LeviMNuclear receptors in renal diseaseBiochim Biophys Acta2011181281061106721511032
  • GardPRImplications of the angiotensin converting enzyme gene insertion/deletion polymorphism in health and disease: a snapshot reviewInt J Mol Epidemiol Genet20101214515721537387
  • Sayed-TabatabaeiFAOostraBAIsaacsAvan DuijnCMWittemanJCACE polymorphismsCirc Res20069891123113316690893
  • KramersCDanilovSMDeinumJPoint mutation in the stalk of angiotensin-converting enzyme causes a dramatic increase in serum angiotensin-converting enzyme but no cardiovascular diseaseCirculation2001104111236124011551873
  • RiederMJTaylorSLClarkAGNickersonDASequence variation in the human angiotensin converting enzymeNat Genet1999221596210319862
  • RuggenentiPBettinaglioPPinaresFRemuzziGAngiotensin converting enzyme insertion/deletion polymorphism and renoprotection in diabetic and nondiabetic nephropathiesClin J Am Soc Nephrol2008351511152518550651
  • SantosPCKriegerJEPereiraACRenin-angiotensin system, hypertension, and chronic kidney disease: pharmacogenetic implicationsJ Pharmacol Sci20121202778823079502
  • PennoGChaturvediNTalmudPJEffect of angiotensin-converting enzyme (ACE) gene polymorphism on progression of renal disease and the influence of ACE inhibition in IDDM patients: findings from the EUCLID Randomized Controlled Trial. EURODIAB Controlled Trial of Lisinopril in IDDMDiabetes1998479150715119726242
  • UedaSMeredithPAMortonJJConnellJMElliottHLACE (I/D) genotype as a predictor of the magnitude and duration of the response to an ACE inhibitor drug (enalaprilat) in humansCirculation19989820214821539815869
  • JacobsenPRossingKRossingPAngiotensin converting enzyme gene polymorphism and ACE inhibition in diabetic nephropathyKidney Int1998534100210069551410
  • JacobsenPTarnowLCarstensenBHovindPPoirierOParvingHHGenetic variation in the Renin-Angiotensin system and progression of diabetic nephropathyJ Am Soc Nephrol200314112843285014569094
  • ParvingHHde ZeeuwDCooperMEACE gene polymorphism and losartan treatment in type 2 diabetic patients with nephropathyJ Am Soc Nephrol200819477177918199798
  • SoWYMaRCOzakiRAngiotensin-converting enzyme (ACE) inhibition in type 2, diabetic patients – interaction with ACE insertion/deletion polymorphismKidney Int20066981438144316395257
  • FilighedduFArgiolasGBullaEClinical variables, not RAAS polymorphisms, predict blood pressure response to ACE inhibitors in SardiniansPharmacogenomics20089101419142718855530
  • YuHZhangYLiuGRelationship between polymorphism of the angiotensin-converting enzyme gene and the response to angiotensin-converting enzyme inhibition in hypertensive patientsHypertens Res2003261188188614714579
  • ArnettDKDavisBRFordCEPharmacogenetic association of the angiotensin-converting enzyme insertion/deletion polymorphism on blood pressure and cardiovascular risk in relation to antihypertensive treatment: the Genetics of Hypertension-Associated Treatment (GenHAT) studyCirculation2005111253374338315967849
  • NonoguchiHNakayamaYShiigaiTLow-responders to angiotensin II receptor blockers and genetic polymorphism in angiotensin-converting enzymeClin Nephrol200768420921517969487
  • AdlerSRenal disease: environment, race, or genes?Ethn Dis2006162 Suppl 2S2-35-3916774008
  • KearneyPMWheltonMReynoldsKMuntnerPWheltonPKHeJGlobal burden of hypertension: analysis of worldwide dataLancet2005365945521722315652604
  • IbrahimMMDamascenoAHypertension in developing countriesLancet2012380984161161922883510
  • ShawJESicreeRAZimmetPZGlobal estimates of the prevalence of diabetes for 2010 and 2030Diabetes Res Clin Pract201087141419896746
  • WangYMiJShanXYWangQJGeKYIs China facing an obesity epidemic and the consequences? The trends in obesity and chronic disease in ChinaInt J Obes (Lond)200731117718816652128
  • HallMEdo CarmoJMda SilvaAAJuncosLAWangZHallJEObesity, hypertension, and chronic kidney diseaseInt J Nephrol Renovasc Dis20147758824600241
  • KöttgenAPattaroCBögerCANew loci associated with kidney function and chronic kidney diseaseNat Genet201042537638420383146
  • SummersSASteinmetzOMOoiJDToll-like receptor 9 enhances nephritogenic immunity and glomerular leukocyte recruitment, exacerbating experimental crescentic glomerulonephritisAm J Pathol201017752234224420847283
  • SummersSAHoiASteinmetzOMTLR9 and TLR4 are required for the development of autoimmunity and lupus nephritis in pristane nephropathyJ Autoimmun201035429129820810248
  • BochudPYHersbergerMTaffePPolymorphisms in Toll-like receptor 9 influence the clinical course of HIV-1 infectionAIDS200721444144617301562
  • KrayenbuehlPAHersbergerMTruningerKToll-like receptor 4 gene polymorphism modulates phenotypic expression in patients with hereditary hemochromatosisEur J Gastroenterol Hepatol201022783584119809335
  • YangHYLuKCLeeHSRole of the functional Toll-Like receptor-9 promoter polymorphism (−1237T/C) in increased risk of end-stage renal disease: a case-control studyPLoS One201383e5844423472199
  • Jimenez-SousaMALópezEFernandez-RodriguezAGenetic polymorphisms located in genes related to immune and inflammatory processes are associated with end-stage renal disease: a preliminary studyBMC Med Genet2012135822817530
  • SinghVJaiswalPKTiwariPKapoorRMittalRDAssociation of chemokine gene variants with end stage renal disease in North Indian populationTranspl Immunol201328418919223615182
  • NabrdalikKGumprechtJAdamczykPGórczyńska-KosiorzSZywiecJGrzeszczakWAssociation of rs1800471 polymorphism of TGFB1 gene with chronic kidney disease occurrence and progression and hypertension appearanceArch Med Sci20139223023723671432
  • BuraczynskaMBaranowicz-GaszczykIBorowiczEKsiazekATGF-beta1 and TSC-22 gene polymorphisms and susceptibility to microvascular complications in type 2 diabetesNephron Physiol20071064p69p7517622752
  • CollECormandBCamposBAssociation of TGF-beta1 polymorphisms with chronic renal diseaseJ Nephrol200417679479915593053
  • NgDPWarramJHKrolewskiASTGF-beta 1 as a genetic susceptibility locus for advanced diabetic nephropathy in type 1 diabetes mellitus: an investigation of multiple known DNA sequence variantsAm J Kidney Dis2003411222812500218
  • McKnightAJSavageDAPattersonCCSadlierDMaxwellAPResequencing of genes for transforming growth factor beta1 (TGFB1) type 1 and 2 receptors (TGFBR1, TGFBR2), and association analysis of variants with diabetic nephropathyBMC Med Genet20078517319955
  • BabelNGabdrakhmanovaLHammerMHPredictive value of cytokine gene polymorphisms for the development of end-stage renal diseaseJ Nephrol200619680280717173255
  • YuZYChenLSZhangLCZhouTBMeta-analysis of the relationship between ACE I/D gene polymorphism and end-stage renal disease in patients with diabetic nephropathyNephrology (Carlton)201217548048722385293
  • ZhouTBYinSSQinYHAssociation of angiotensinogen M235T gene polymorphism with end-stage renal disease risk: a meta-analysisMol Biol Rep201340276577223065231
  • SilvaPSFontanaVLuizonMReNOS and BDKRB2 genotypes affect the antihypertensive responses to enalaprilEur J Clin Pharmacol201369216717722706620
  • BeharDMRossetSTzurSAfrican ancestry allelic variation at the MYH9 gene contributes to increased susceptibility to non-diabetic end-stage kidney disease in Hispanic AmericansHum Mol Genet20101991816182720144966
  • FreedmanBIHicksPJBostromMANon-muscle myosin heavy chain 9 gene MYH9 associations in African Americans with clinically diagnosed type 2 diabetes mellitus-associated ESRDNephrol Dial Transplant200924113366337119567477
  • FreedmanBIHicksPJBostromMAPolymorphisms in the non-muscle myosin heavy chain 9 gene (MYH9) are strongly associated with end-stage renal disease historically attributed to hypertension in African AmericansKidney Int200975773674519177153
  • KaoWHKlagMJMeoniLAMYH9 is associated with nondiabetic end-stage renal disease in African AmericansNat Genet200840101185119218794854
  • KoppJBSmithMWNelsonGWMYH9 is a major-effect risk gene for focal segmental glomerulosclerosisNat Genet200840101175118418794856
  • CookeJNBostromMAHicksPJPolymorphisms in MYH9 are associated with diabetic nephropathy in European AmericansNephrol Dial Transplant20122741505151121968013
  • MatshaTEMasconiKYakoYYPolymorphisms in the non-muscle myosin heavy chain gene (MYH9) are associated with lower glomerular filtration rate in mixed ancestry diabetic subjects from South AfricaPLoS One2012712e5252923285077
  • TaviraBCotoEGómezJAssociation between a MYH9 polymorphism (rs3752462) and renal function in the Spanish RENASTUR cohortGene20135201737623470845
  • FreedmanBIKoppJBLangefeldCDThe apolipoprotein L1 (APOL1) gene and nondiabetic nephropathy in African AmericansJ Am Soc Nephrol20102191422142620688934
  • GenoveseGFriedmanDJRossMDAssociation of trypanolytic ApoL1 variants with kidney disease in African AmericansScience2010329599384184520647424
  • GenoveseGTonnaSJKnobAUA risk allele for focal segmental glomerulosclerosis in African Americans is located within a region containing APOL1 and MYH9Kidney Int201078769870420668430
  • TzurSRossetSShemerRMissense mutations in the APOL1 gene are highly associated with end stage kidney disease risk previously attributed to the MYH9 geneHum Genet2010128334535020635188
  • O’SeaghdhaCMParekhRSHwangSJThe MYH9/APOL1 region and chronic kidney disease in European-AmericansHum Mol Genet201120122450245621429915
  • WitaspANordforsLCarreroJJGenetic studies in chronic kidney disease: interpretation and clinical applicabilityJ Nephrol201225685186423042432
  • PriorTWCarrier screening for spinal muscular atrophyGenet Med2008101184084218941424
  • MillerDTAdamMPAradhyaSConsensus statement: chromosomal microarray is a first-tier clinical diagnostic test for individuals with developmental disabilities or congenital anomaliesAm J Hum Genet201086574976420466091
  • RobsonMOffitKClinical practice. Management of an inherited predisposition to breast cancerN Engl J Med2007357215416217625127
  • FruehFWAmurSMummaneniPPharmacogenomic biomarker information in drug labels approved by the United States food and drug administration: prevalence of related drug usePharmacotherapy200828899299818657016
  • WienkersLCHeathTGPredicting in vivo drug interactions from in vitro drug discovery dataNat Rev Drug Discov200541082583316224454
  • MolanaeiHCarreroJJHeimbürgerOInfluence of the CYP2D6 polymorphism and hemodialysis on codeine disposition in patients with end-stage renal diseaseEur J Clin Pharmacol201066326927319940985
  • van EssenGGRensmaPLde ZeeuwDAssociation between angiotensin-converting-enzyme gene polymorphism and failure of renoprotective therapyLancet1996347899494958538349
  • CarreroJJQureshiARPariniPLow serum testosterone increases mortality risk among male dialysis patientsJ Am Soc Nephrol200920361362019144759
  • MuntingheFLVerduijnMZuurmanMWCCR5 deletion protects against inflammation-associated mortality in dialysis patientsJ Am Soc Nephrol20092071641164919389855
  • MuntingheFLVegterSVerduijnMUsing a genetic, observational study as a strategy to estimate the potential cost-effectiveness of pharmacological CCR5 blockade in dialysis patientsPharmacogenet Genomics201121741742521597398
  • BoardPCogganMJohnstonPRossVSuzukiTWebbGGenetic heterogeneity of the human glutathione transferases: a complex of gene familiesPharmacol Ther19904833573692084706
  • RybergDSkaugVHewerAGenotypes of glutathione transferase M1 and P1 and their significance for lung DNA adduct levels and cancer riskCarcinogenesis1997187128512899230269
  • LinYSHungSCWeiYHTarngDCGST M1 polymorphism associates with DNA oxidative damage and mortality among hemodialysis patientsJ Am Soc Nephrol200920240541519056870
  • FiliopoulosVVlassopoulosDInflammatory syndrome in chronic kidney disease: pathogenesis and influence on outcomesInflamm Allergy Drug Targets20098536938220025585
  • NordforsLLindholmBStenvinkelPEnd-stage renal disease – not an equal opportunity disease: the role of genetic polymorphismsJ Intern Med2005258111215953127
  • StenvinkelPChronic kidney disease: a public health priority and harbinger of premature cardiovascular diseaseJ Intern Med2010268545646720809922
  • HunterDJGene-environment interactions in human diseasesNat Rev Genet20056428729815803198
  • SmildeTDAsselbergsFWHillegeHLMild renal dysfunction is associated with electrocardiographic left ventricular hypertrophyAm J Hypertens200518334234715797651
  • SmildeTDZuurmanMWHillegeHLRenal function dependent association of AGTR1 polymorphism (A1166C) and electrocardiographic left-ventricular hypertrophyAm J Hypertens200720101097110317903694
  • KuznetsovaTStaessenJAThijsLLeft ventricular mass in relation to genetic variation in angiotensin II receptors, renin system genes, and sodium excretionCirculation2004110172644265015492316
  • OhishiMRakugiHOgiharaTAssociation between a deletion polymorphism of the angiotensin-converting-enzyme gene and left ventricular hypertrophyN Engl J Med199433116109710987993479
  • SchunkertHControversial association of left ventricular hypertrophy and the ACE I/D polymorphism – is the mist clearing up?Nephrol Dial Transplant1998135110911129623534
  • BrewsterUCPerazellaMAThe renin-angiotensin-aldosterone system and the kidney: effects on kidney diseaseAm J Med2004116426327214969655
  • van der KleijFGSchmidtANavisGJAngiotensin converting enzyme insertion/deletion polymorphism and short-term renal response to ACE inhibition: role of sodium statusKidney Int Suppl199763S23S269407415
  • DwivediRSHermanJGMcCaffreyTARajDSBeyond genetics: epigenetic code in chronic kidney diseaseKidney Int2011791233220881938
  • StenvinkelPKarimiMJohanssonSImpact of inflammation on epigenetic DNA methylation – a novel risk factor for cardiovascular disease?J Intern Med2007261548849917444888
  • HarveySJJaradGCunninghamJPodocyte-specific deletion of dicer alters cytoskeletal dynamics and causes glomerular diseaseJ Am Soc Nephrol200819112150215818776121
  • HoJJMarsdenPADicer cuts the kidneyJ Am Soc Nephrol200819112043204618923053
  • ShiSYuLChiuCPodocyte-selective deletion of dicer induces proteinuria and glomerulosclerosisJ Am Soc Nephrol200819112159216918776119
  • HoJNgKHRosenSDostalAGregoryRIKreidbergJAPodocyte-specific loss of functional microRNAs leads to rapid glomerular and tubular injuryJ Am Soc Nephrol200819112069207518832437
  • PernaAFIngrossoDSattaELombardiCAcanforaFDe SantoNGHomocysteine metabolism in renal failureCurr Opin Clin Nutr Metab Care200471535715090904
  • KalluriRNeilsonEGEpithelial-mesenchymal transition and its implications for fibrosisJ Clin Invest2003112121776178414679171
  • RashidAIssaJPCpG island methylation in gastroenterologic neoplasia: a maturing fieldGastroenterology200412751578158815521024
  • BechtelWMcGoohanSZeisbergEMMethylation determines fibroblast activation and fibrogenesis in the kidneyNat Med201016554455020418885
  • ParsaAKaoWHXieDAPOL1 risk variants, race, and progression of chronic kidney diseaseN Engl J Med2013369232183219624206458