Skip to main content
American Journal of Physiology - Renal Physiology logoLink to American Journal of Physiology - Renal Physiology
. 2016 Dec 14;312(3):F375–F384. doi: 10.1152/ajprenal.00266.2016

A perspective on chronic kidney disease progression

Jianyong Zhong 1,2,*, Hai-Chun Yang 1,2,*, Agnes B Fogo 1,2,3,
PMCID: PMC5374308  PMID: 27974318

Abstract

Chronic kidney disease (CKD) will progress to end stage without treatment, but the decline of renal function may not be linear. Compared with glomerular filtration rate and proteinuria, new surrogate markers, such as kidney injury molecule-1, neutrophil gelatinase-associated protein, apolipoprotein A-IV, and soluble urokinase receptor, may allow potential intervention and treatment in the earlier stages of CKD, which could be useful for clinical trials. New omic-based technologies reveal potential new genomic and epigenomic mechanisms that appear different from those causing the initial disease. Various clinical studies also suggest that acute kidney injury is a major risk for progressive CKD. To ameliorate the progression of CKD, the first step is optimizing renin-angiotensin-aldosterone system blockade. New drugs targeting endothelin, transforming growth factor-β, oxidative stress, and inflammatory- and cell-based regenerative therapy may have add-on benefit.

Keywords: surrogate marker, genomic, acute kidney injury, regenerative therapy


the prevalence of chronic kidney disease (CKD) is estimated to be 8–16% worldwide (38). In patients over 64 yr old, the prevalence increases to 23.4–35.8%, suggesting increasing age contributes to increased CKD (104). The yearly economic costs of care for CKD and end-stage renal disease (ESRD) in patients over age 65 are $60 billion, representing 24% of total Medicare expenditures in 2011 in the United States of America. The diagnostic criteria for CKD are: a glomerular filtration rate (GFR) threshold <60 ml·min−1·1.73 m−2 or the presence of kidney damage ≥3 mo. Kidney damage refers to pathological abnormalities documented by biopsy or imaging, alterations in urinary sediment or proteinuria (urine protein-to-creatinine ratio >200 mg/g or urine albumin-to-creatinine >30 mg/g), genetic disorders, or a history of renal transplantation (47). In 2002, the National Kidney Foundation Kidney Disease Outcomes Quality Initiative classified CKD into five stages according to estimated GFR (eGFR) (60). These guidelines in 2012 recommended classification of CKD based not only on the cause and GFR category but also albuminuria category because of the graded relationship between increasing proteinuria and a variety of important outcomes, including all-cause mortality, cardiovascular disease, and kidney failure (35). The overall awareness of CKD is remarkably low: 90% of individuals with two to four CKD markers and 84% of people with more than five CKD markers were reported to be unaware of their kidney disease (87). Although it is still debated whether early screening for CKD should be done in the population, early stage intervention treatment is important to improve the quality of life and survival (50). Numerous experimental models attempt to model the human disease conditions, but none completely capture the mechanisms and phenotypes of human CKD. In this regard, we quote Pickering et al. who eloquently stated: “We would make a plea that so far as any conclusions are drawn as to the mechanism of human disease, the evidence derived from man should at least be considered” (68). This review will therefore focus on new findings from human studies related to detection, mechanism, and treatment of progressive CKD.

Definition of Progressive CKD

Progress rate of CKD.

The progression rate of CKD varies among individuals. Some CKD patients maintain stable eGFR levels over several years, so-called “stable CKD,” or even improve eGFR, so-called “reversal CKD.” In contrast, other CKD patients lose eGFR over time, so-called “progressive CKD.” Progressive CKD is observed in the majority of CKD patients while reversal CKD is uncommon. Follow-up for 12 yr in the African American Study of Kidney Disease and Hypertension (AASK) trial showed that 3.3% patients improved eGFR with a mean slope of +1.06 ml·min−1·1.73 m−2·yr−1 per year compared with −2.45 (0.07) ml·min−1·1.73 m−2·yr−1 among the remaining patients (32). In another small 10-yr follow-up study, 167 of 347 (48.1%) CKD III patients did not progress while 60 (17.3%) progressed to stage 4 and 120 (34.6%) progressed to stage 5 (4). Another study from France showed improved eGFR in 15.3% of patients with a median slope +1.88 ml·min−1·yr−1 even in CKD stage 4–5 (96). Similar results were observed in the Modification of Diet in Renal Disease Study. Stable eGFR was observed in 19% and reversal of CKD in 11% over 2 yr follow-up (33). Significantly improved eGFR slope was detected in 48.2% of CKD stage 2 patients, 29.3% of CKD stage 3 patients, and only 14.7% of CKD stage 4 patients (86). Clearly, whether CKD progresses or not is related to the CKD stage at enrollment and beginning of intervention.

Nonlinear pattern for progressive CKD.

The progression of CKD may not be linear. CKD progression, especially GFR decline, has been assumed to follow a linear or possibly a loglinear trajectory. Physicians have used the linear decline model to counsel patients on when they might reach ESRD and need renal replacement therapy (33, 58). Newer data suggest that the natural pattern of progression from CKD to ESRD followed a more staccato and unpredictable course (74, 103). In a study of individual GFR progression trajectories over 12 yr of follow-up, Li et al. demonstrated that 41.6% of CKD patients showed a >0.9 probability of having either a nonlinear trajectory or a prolonged nonprogression period while in 66.1% of patients the probability of these nonlinear courses was >0.5 (51).

Markers for progressive CKD.

To predict and define progressive CKD is still difficult, especially with short follow-up time. This is related to a lack of a consensus definition for CKD progression and lack of sensitive and specific biomarkers for the early prediction of CKD progression. True end points for CKD, such as development of ESRD, which is often defined as a new initiation of renal replacement therapy, may not be reached for decades. Surrogate end points have potential advantages over true clinical end points, which could reduce the cost and offer more opportunities for clinical trials (Table 1). The most popular surrogate markers for CKD are albuminuria, serum creatinine, and GFR. As an early marker of renal damage, microalbuminuria may allow earlier interventions for CKD (48). However, it is unclear whether reducing microalbuminuria is necessary for inhibiting CKD progression and improving clinical outcomes (26). Another accepted surrogate end point for progression of CKD to ESRD is doubling of serum creatinine and/or 50% reduction of GFR. Doubling of serum creatinine corresponds to a 57% change in eGFR. In acute kidney injury (AKI), serum creatinine has been found as a late and often insensitive marker of underlying injury. Among CKD patients with baseline eGFR <60 ml·min−1·1.73 m−2, the adjusted hazard ratios for ESRD were 32.1 for changes of −57% in eGFR and 5.4 for changes of −30%. Average adjusted 10-yr risk of ESRD was 99% for eGFR change of −57%, was 83% for eGFR change of −40%, and was 64% for eGFR change of −30% vs. 18% for eGFR change of 0%. Corresponding mortality risks were 77, 60, and 50% vs. 32%, showing a similar but weaker pattern (17). This and other studies suggested using eGFR declines of 30 and 40% as alternative surrogate end points of progression, which may offer the advantage of being earlier and more common markers of deteriorating kidney function, potentially allowing shorter clinical trial duration.

Table 1.

Surrogate markers for progressive CKD

Marker CKD Stages Advantage Disadvantage Ref. No.
Standard markers Albuminuria 1–3 Sensitive Not correlated to CKD progression (26, 48)
Doubling of serum creatinine 2–4 Correlated to CKD progression Insensitive (17)
50% reduction of GFR 2–4 Correlated to CKD progression Insensitive (17)
Novel Biomarkers KIM-1 1–3 Sensitive, correlated with eGFR decline Need to validate threshold (72)
NGAL 1–3 Sensitive, correlated with eGFR decline Need to validate threshold (6, 8, 78)
ApoA-IV 1–3 Sensitive, correlated with eGFR decline Need to validate for threshold (7)
suPAR 1–3 Sensitive, correlated with eGFR decline Need to validate for threshold (29)

CKD, chronic kidney disease; GFR, glomerular filtration rate; KIM-1, kidney injury molecule-1; NGAL, neutrophil gelatinase-associated protein; ApoA-IV, apolipoprotein A-IV; suPAR, soluble urokinase receptor; eGFR, estimated GFR.

Some biomarkers can be considered to be intermediate end points. Currently, the use of biomarkers for clinical decision making is not defined, but some markers, such as kidney injury molecule (KIM-1), neutrophil gelatinase-associated protein (NGAL), apolipoprotein A-IV (apoA-IV), and soluble urokinase receptor (suPAR), appear to be good candidates. In a retrospective analysis of 107 diabetic type 1 with CKD stages 1–3 followed for 5–15 yr, 63% of those subjects with higher KIM-1 levels (>97 pg/ml) progressed to ESRD, whereas only 20% of patients with lower levels progressed. In addition, baseline plasma KIM-1 levels correlated with rate of eGFR decline after adjustment for baseline urinary albumin-to-creatinine ratio, eGFR, and HbA1c (72). As an established marker for AKI, NGAL has also been associated with CKD incidence and progression in adults. In a community-based study, NGAL was evaluated as an independent risk factor for incident CKD. Participants with NGAL concentrations in the highest quartile had more than twofold higher odds of incident CKD stage 3 compared with those with NGAL in the lowest quartile after multivariable adjustment. Adjustment for urinary creatinine and albumin concentration attenuated this association (6). In a cohort of 158 elderly Caucasian predialysis CKD patients with low-grade proteinuria, urinary NGAL-to-creatinine ratio was associated with mortality and renal replacement therapy, and this risk was independent of kidney and cardiovascular risk factors (78). Similar results were found in a cohort of 96 CKD patients followed for 18.5 mo, where urinary NGAL and sNGAL predicted CKD progression independently of other potential confounders, including eGFR and age (8). Human apoA-IV is a 46-kDa glycoprotein synthesized in intestinal enterocytes during fat absorption and incorporated on the surface of chylomicrons. Increased baseline ApoA-IV levels were found in those mild to moderate CKD patients who progressed over 7 yr follow-up in a small study of 177 patients. Serum ApoA-IV increase by 1 mg/dl predicted progression, with 11 ml/min decrease in GFR, with an area under the curve of 0.792 (P < 0.001) and a hazard ratio of 1.062 (P = 0.006) (7). In the Emory Cardiovascular Biobank cohort and the Women’s Interagency HIV Study cohort, a higher suPAR level at baseline was associated with a greater decline in the eGFR. The participants with a normal eGFR at baseline had the largest suPAR-related decline in the eGFR (29). The above biomarkers need to be validated to identify thresholds and cut-offs for prediction of CKD progression and adverse events by additional large multicenter prospective studies. Additional studies are also required to determine whether the biomarkers continue to predict CKD progression longitudinally, in addition to merely associating with the baseline levels of GFR that correlate with progression. It is likely that a panel of CKD biomarkers will provide more information than any one alone. Such a panel may need to be context-specific based on pathophysiological considerations. For example, distinct panels may emerge for prediction of CKD due to etiology, such as diabetes and primary glomerulonephritis vs. reflecting the major underlying pathological feature such as tubulointerstitial fibrosis and inflammation. Ongoing discoveries using techniques such as proteomics, peptidomics, urinary transcriptomics, and micro-RNA analysis are continuing to reveal novel biomarkers and therapeutic targets. The CKD Biomarkers Consortium has 15 ongoing studies with the aim to develop and validate novel biomarkers for CKD.

New Insights into Mechanisms of CKD Progression

Progressive CKD may be viewed as having three phases. First, there is cause-specific injury and acute response to that injury. In the second phase, misdirected repair generates fibrosis and dysfunction. At this phase, although fibrosis is a pathological and destructive event, it is essentially a self-limiting repair process to restrict the injury. The third and final stage is that of relatively steady progressive loss of remnant nephrons, which requires multiple nascent injury to each nephron or cluster of nephrons. Thus, the causes of CKD incidence appear different from those driving CKD progression, since progression rates of CKD differ dramatically among patients with apparent identical primary diseases (71).

Genetic and epigenetic variants.

Genome-wide association studies (GWAS) focus on the most common genetic variations in the human genome. Single nucleotide polymorphisms (SNPs) are common substitutions of a single base with another, which occur with high frequency in the human genome (1 per 300–500 base pairs) (36). Although most SNPs have no functional outcome, some might result in biological changes, which could play a role in disease susceptibility. Compared with some rare diseases that are caused by single locus mutations, the genetic component of common polygenic diseases, such as CKD, is thought to involve many common genetic variants (34). Because a single SNP explains only a small proportion of a trait’s variance, multiple genetic variants are required to account for the total genetic risk of a disease. GWAS identified several genes, in which variants are associated with decline of renal function in CKD, including uromodulin (UMOD), nonmuscle myosin heavy chain type 2 isoform A, methenyltetrahydrofolate synthetase, eyes absent homolog 1, and transcription factor-7-like 2 (43, 45, 67). In a follow-up analysis, the presence of the UMOD SNP rs4293393 was found to be associated with uromodulin levels, and elevated uromodulin levels preceded the development of CKD (44). Apolipoprotein L1 (APOL1)-mediated risk for CKD progression was also reported by the AASK and Chronic Renal Insufficiency Cohort (CRIC) studies (52, 65). These longitudinal studies observed that the presence of two APOL1 risk variants was associated with more rapid loss of kidney function in the nondiabetic subjects in AASK and in both diabetic and nondiabetic subjects in CRIC. The rates of CKD progression were lowest for European Americans (who essentially lack APOL1 risk variants), intermediate for African Americans with zero or one APOL1 risk variant, and highest for African Americans with two APOL1 risk variants (65). Interactions between APOL1 and several modifiable environmental factors, or between different genes, produce the variable clinical phenotypes, which show different response to conventional therapies targeting reductions in systemic blood pressure and proteinuria.

Protein- or mRNA-based biomarkers often provide a snap-shop at time of analysis over a long-term picture, which underlies individual progression of CKD. Recent studies suggest that these limitations can be overcome by analysis of epigenetic markers, because epigenetics in general offer the advantage of greater stability but do underlie modifications of gene expression during disease progression (82). Unlike genetic polymorphisms, epigenetic modifications can be more easily therapeutically modified. There are three major epigenetic mechanisms, namely DNA methylation, micro-RNAs, and histone modifications, which interact and impact each other. DNA methylation refers to clustering of methylated cytosine bases within a specific promoter region (CpG island promoters) (101). In the CRIC study, which was established to follow a diverse group of patients with chronic renal insufficiency with intensive screening and follow-up for the purpose of identifying high-risk groups, patients characterized as rapid progressors had different DNA methylation profiles of a number of genes that have been implicated in inflammation, oxidative stress, or fibrosis (97). Altered presence of a single micro-RNA causes altered expression of numerous genes, typically requiring additional transcriptional profiling to assess context-dependent relevance (84). Renal biopsy specimens from patients with so-called “hypertensive nephrosclerosis” have enrichment of miR-200a, miR-200b, miR-141, miR-429, miR-205, and miR-192 expression, and the degree of upregulation correlated with disease severity (94). Histone modifications are most complex, since those occur at multiple sites within multiple genes, blurring assessment of biological impact of identified modification (83). Transforming growth factor-β (TGF-β) increases histone H3 lysine methylation (H3K4me1, H3K4me2, and H3K4me3), which increased expression of connective tissue growth factor (CTGF), a downstream effector of TGF-β’s profibrotic effects, collagen-1α1, and plasminogen activator inhibitor-1, which inhibits fibrinolysis and proteolysis in mesangial cells (81). Blocking class I histone deacetylatase through the selective class I histone deacetylase inhibitor MS-275 led to inhibition of TGF-β signaling and blocked renal fibroblast activation (53).

AKI and CKD.

Several studies indicate that AKI has a deleterious long-term effect on the morbidity and mortality of patients (12, 55). As a significant risk factor, AKI induced an 8.8-fold increase in risk for CKD and a 3.3-fold increase in risk for ESRD (16). One study showed acute protective effects by a p53 inhibitor in an ischemia-reperfusion model but worsened fibrosis after 8 wk, indicating different mechanism in AKI vs. AKI-to-CKD transition (18). The degree of increase in serum creatinine during the AKI episode has been linked to increased subsequent development of ESRD (11). The severity, duration, and frequency of episodes of AKI are now recognized as key determinants influencing progression to CKD. Kidney fibrosis probably starts as a beneficial reparative mechanism in response to an initial damage. If one or more of the initial stages of this process are not correctly regulated, a pathological fibrosis is originated and progresses to CKD. The maladaptive repair to AKI represents a necessary but not sufficient ingredient to promote progression (92). The mechanism by which AKI leads to CKD is unclear, but several mechanisms have been proposed, such as nephron loss, inflammation, endothelial injury with vascular rarefaction and hypoxia, as well as epigenetic changes and cell cycle arrest in epithelial cells (3, 12, 100). For instance, vascular rarefaction of peritubular capillaries is correlated with the severity of fibrosis and predicts both interstitial damage and decreased GFR (15). Furthermore, many experimental findings in rodents strongly suggest that vascular rarefaction contributes to the decrease of GFR and the progression of CKD (41).

AKI not only can transform into CKD but also contribute to the progression of CKD from other causes. Brenner and colleagues showed that glomerular hypertension and hyperfiltration are major factors accounting for progression of CKD in animal models and humans (10). However, tubular atrophy and interstitial fibrosis are the common end points of practically all progressive kidney diseases, irrespective of the initial etiology, and cardinal features of CKD (9). Studies indicated that interstitial scarring may not be directly progressive in nature, but it exaggerates responses to secondary injury, reduces renal functional reserve, and contributes to the development of hypertension (39, 91). Broadly, a vicious cycle of “glomerulus-tubulointerstitial-glomerulus” injury can promote CKD progression. Interdicting any feedback in this cycle, for example, by reducing hypoxia or interstitial inflammation, could contribute to slow or stop the progression of CKD. The clinical follow-up of survivors of AKI is low, which may result in missed opportunity to prevent chronic disease (28).

New Treatment Approaches for Progressive CKD

The primary aims when treating CKD are both to slow the progression of CKD and to prevent cardiovascular disease, the principal cause of morbidity and mortality in the CKD population (93). The main approaches to slowing the rate of CKD progression are treatment of the underlying disease, if possible; treatment of reversible causes of renal failure, which, if identified and corrected, may result in the recovery of renal function; and treatment of secondary factors that are predictive of progression, such as elevated blood pressure and proteinuria, when the renal damage has already occurred. Furthermore, strong evidence from murine studies suggest that renal fibrosis in principle is a treatable target and that possible regression of fibrosis would translate into preservation of kidney function, although regression of CKD without intervention is rare in humans. The concept of regenerative nephrology is just now emerging, focusing on two key ideas. Kidney regeneration could be achieved through use of growth factors and morphogens, or multipotent cells could be “taught” to regenerate the chronically injured kidney. Both concepts require either recreation of a growth factor environment within the kidney to facilitate renal regeneration or generation of renal cell type-specific progenitor cells in vitro to repopulate the kidney. There are several common concepts related to standard and regenerative medical therapies of CKD (Table 2).

Table 2.

Approaches for CKD treatment

Strategy Drug Name Ref. No.
Optimize and maximize RAAS blockade RAAS blocker Renin inhibitor, ACEI, ARB, Aldosterone synthesis inhibitor (25, 57, 66, 73, 77)
New targets ETAR antagonist Atrasentan (21, 42)
TGF-β inhibitor Pirfenidone (14, 75)
Antioxidant Allopurinol, Febuxostat (27, 31, 40, 54)
Anti-inflammation CCX140, Pentoxifylline (20, 61)
SGLT2 inhibitor Empagliflozin (95)
Regenerative medicine Stimulate renal regeneration BMP-7 (59, 80, 102)
Stem cell-based regenerative therapy HSCs, MSCs, EPCs (2, 23, 37, 64, 69, 76, 79, 85)

RAAS, renin-angiotensin-aldosterone system; ACEI, angiotensin-converting enzyme inhibitor; ARB, ANG II receptor blocker; ETAR, endothelin receptor type A; TGF-β, transforming growth factor-β; SGLT2, sodium glucose cotransporter 2; BMP-7, bone morphogenetic protein-7; HSCs, hematopoietic stem cells; MSCs, mesenchymal stem cells; EPCs, endothelial progenitor cells.

Optimize and maximize renin-angiotensin-aldosterone system blockade.

Current renoprotection paradigms generally depend on the use of angiotensin-converting enzyme inhibitor (ACEI) and/or ANG II receptor blockers (ARBs), which have been shown to reduce proteinuria and retard the progression of CKD. However, ANG II and aldosterone levels increase after chronic ACEI or ARBs treatment, so-called ANG II-escape and aldosterone escape. A reactive rise in renin levels occurs when mineralcorticoid receptor antagonists or renin inhibitors are used. Chronically increased ANG II and aldosterone worsens diseases such as heart failure and renal disease (73, 77). These compensatory responses at different levels prove that single renin-angiotensin-aldosterone system (RAAS) blocker cannot provide full blockade of the RAAS cascade, suggesting dual therapy may have more benefit. However, kidney outcomes with telmisartan, ramipril, or both, in people at high vascular risk (ONTARGET), and combined angiotensin inhibition for the treatment of diabetic nephropathy (VA-Nephron-D trial), unexpectedly showed increased risk of adverse outcomes and events, such as AKI, hyperkalemia, and/or need for dialysis (25, 57). The combination therapy of a renin inhibitor (aliskerin) along with an ARB or an ACEI in type 2 diabetes (ALTITUDE) showed similar increased risks of cardiovascular events (66). Despite this side effect, dual blockade reduced proteinuria and decreased ESRD events compared with monotherapy. In animal studies, combination of an aldosterone synthesis inhibitor and ARB showed more renal benefit with low risk of hyperkalemia, suggesting this combination could be an additional choice for RAAS. Another consideration is that these dual therapy approaches used recommended doses for monotherapy for both drugs. A different therapeutic strategy based on the combination of lower than recommended doses for monotherapy of an ACEI and an ARB has been suggested to effectively block the renin-angiotensin system without excess blood pressure reduction and side effects. Now, the ongoing VALID trial is testing whether halved doses of an ACEI and an ARB may be more effective than full doses of each agent alone in diabetic nephropathy (ClinicalTrials.gov: NCT00494715). In this case, the dual therapy could be considered for patients with residual proteinuria despite maximal monotherapy of RAAS blockade, with close monitoring of blood pressure, heart and renal function.

New targets for CKD.

Considering the incomplete efficacy of RAAS blockade, it is necessary to find new drugs that could either exert a complementary action to ACEI and ARBs or act on other pathophysiological processes involved in the progression of CKD. The great expectations of novel drug therapies for CKD management over the last decade have not come to fruition. Several recent candidates have failed to show improved outcome for therapy of diabetic nephropathy because of the following three major themes: 1) insufficient studies of preclinical models to support efficacy or explore potential toxicity, such as the BEACON (bardoxolone, a Nrf2 inducer) and ASCEND [avosentan, endothelin (ET) receptor blocker] studies (19, 56), 2) lack of benefit, such as the SUN-micro (sulodexide) study (63), and 3) sponsors' decisions to discontinue therapeutic development because of business and/or regulatory considerations, such as ruboxistaurin (PKC-β inhibitor) and FG-3019 (anti-CTGF antibody) (1, 88). However, some ongoing clinical trials still offer promise of further gains.

Plasma ET-1 is increased in CKD patients and correlates with urinary albumin excretion and renal function. ET-1 affects renal and extrarenal via activation of two receptor subtypes: endothelin receptor type A and B (ETAR, ETBR). ETAR activation promotes podocyte and mesangial dysfunction, renal inflammation, and oxidative stress, leading to proteinuria and glomerulosclerosis (62). Inhibition of ETBR may induce fluid overload and body weight increase. Although the study for avosentan failed, atrasentan, a more selective ETAR antagonist (ETAR-ETBR blockade 1,800:1 vs. 50-300:1 for avosentan), was studied (Reducing Residual Albuminuria in Subjects with Diabetes and Nephropathy with atrasentan; the RADAR study). A total of 211 subjects with type 2 diabetes mellitus and kidney disease who were on maximum ACEI or ARBs received either 0.75 or 1.25 mg/day of atrasentan or placebo for 12 wk. Albuminuria was maximally reduced by 35 and 38% in the 0.75 and 1.25 mg/day groups, respectively (21). There was no appreciable increase in edema over that of placebo with low-dose atrasentan therapy, suggesting that a renoprotective effect can be obtained in the absence of clinically significant fluid retention. Currently, a large phase 3 trial [Study of Diabetic Nephropathy with Atrsentan (SONAR)] is underway. The study, with a projected enrollment of over 4,000 subjects, will evaluate the effects of atrasentan compared with placebo on cardiovascular morbidity and mortality, urine albumin excretion, changes in eGFR, and impact on quality of life (42).

Pirfenidone is an oral compound with antifibrotic properties. Although its mechanism of action is not fully understood, it inhibits production and activity of TGF-β. The major clinical trial on this topic was performed on 77 patients with diabetic nephropathy (75). After 54 wk, there was a significant improvement in eGFR in patients receiving 1,200 mg of pirfenidone while no statistically significant differences were found in the group receiving a higher dose or in the placebo group. Moreover, no statistical differences were found in the secondary end points: proteinuria and urinary TGF-β level. This failure to decrease albuminuria was also observed in a recent open-label clinical study of pirfenidone in patients with advanced focal segmental glomerulosclerosis, suggesting that the treatment was associated with a reduction in the rate of renal function decline but without attenuating albuminuria (14). These phase II studies could be interpreted as showing a hemodynamic-based effect on eGFR, or, alternatively, the improved eGFR could reflect structural improvement in injury but without change in proteinuria. The latter possibility would be quite different from the current understanding of linkage of benefit of effects on progressive scarring and proteinuria.

Oxidative stress is a contributor to tissue injury in CKD. GKT-137831, a NADPH oxidase 1/4 inhibitor, shows negative data in a phase 2 trial. In contrast, inhibition of xanthine oxidase is more promising. Allopurinol has already shown efficacy in preventing vascular events and slowing kidney function loss in several clinical trials (27, 40). The ongoing clinical trials, PEARL and FEATHER, are currently investigating the specific usefulness of anti-xanthine oxidase (allopurinol and febuxostat) in CKD (31, 54).

Chemokine receptors, such as CCR2, are main drivers of monocyte and macrophage recruitment in diseased kidney. Various resident cells also express CCR2, which drives some of the renal impairment in CKD. Levels of monocyte chemoattractant protein-1, the main ligand for CCR2, are elevated in the kidneys of patients with kidney disease, suggesting reduction of chemokine production is a potential treatment target in CKD. Although a phase II trial of CCR 2/5 antagonists (PF-04634817) failed, another recently completed phase II trial in diabetic nephropathy provided promising results for CCX140, an oral small molecule inhibitor of CCR2. Treatment with CCX140 (5 mg/day) added to standard of care treatment (an ACEI or ARB) resulted in 24% decreasing in albuminuria and the slope of eGFR loss, beyond that achieved with standard of care alone over a 52-wk treatment period. The results of a phase III trial, however, did not confirm any significant impact on GFR but did confirm the anti-proteinuric effect in response to CCX140 (20). Another anti-inflammatory drug, pentoxifylline, showed a slower rate of eGFR loss together with the significant reduction in urine protein excretion when it was added to maximal renin-angiotensin system blockade in CKD patients (61).

The most recent remarkable additional benefits on top of standard RAAS blockade of sodium glucose cotransporter 2 inhibition by empagliflozin in type 2 diabetic patients with high cardiovascular risk to markedly decrease diabetic nephropathy progression are not directly explained by improved glucose control (95). Mechanisms may involve changes in tubuloglomerular feedback, with beneficial intrarenal hemodynamic changes, or other changes in tubular work load, oxidative stress, or other injuries. Thus, one could speculate that benefits may even occur in nondiabetic patients.

Regenerative medicine.

Renal regeneration after acute injury is associated with increased expression of mediators of kidney development, such as Pax-2, Pax-8, Wnt-4, and Wnt-9b, whereas impaired regenerative capacity of the chronically injured kidney is associated with decreased expression of these mediators. A growth factor microenvironment reminiscent of the fetal kidney could be created through administration of selected recombinant growth factors. Bone morphogenetic protein (BMP)-7 is an essential morphogen during kidney development that remains highly expressed in adult kidney (22). Chronic kidney injury is associated with suppression of BMP-7 expression, and supplementation with exogenous recombinant BMP-7 not only inhibits progression of experimental kidney fibrosis but also facilitates reversal of established fibrotic lesions in mice (59, 80, 102). Small-molecule mimetics of BMP-7 are currently undergoing clinical testing.

Cell-based regenerative therapy is being extensively evaluated as an alternative treatment modality. The chief cell types under investigation are hematopoietic stem cells (HSCs), mesenchymal stem cells (MSCs), and endothelial progenitor cells (EPCs). CKD is characterized by reduced renal regenerative capacity with impaired EPC number and function, but there is conflicting evidence regarding MSC functionality and vitality (13, 46, 70, 98). Several studies suggest beneficial regenerative effects of cell-based therapies in animal models of CKD (24, 89, 105). A systematic review and meta-analysis of 71 articles of various animal models found that cell-based therapy reduced development and progression of CKD with decreased urinary protein and plasma urea levels (64). MSCs are being used in several clinical trials in kidney transplant recipients with the aim of increasing immunosuppression and improving regeneration (69, 85). However, the majority of the clinical studies using HSC therapy are focused on lupus nephritis and are all nonrandomized and uncontrolled studies (2, 23, 37, 79). It is noteworthy that several renal diseases, including thrombotic microangiopathy and calcineurin inhibitor nephrotoxicity, can develop after HSC transplantation (76). Drug treatment to indirectly manipulate cell function could also be considered for repopulating progenitor/stem cells. Several randomized trials showed a statin-induced increase in circulating EPC number ranging from 25.8 to 223.5% (30). Several other drugs also affect EPCs, including erythropoietin-stimulating agents, calcium channel blockers, biguanides without or with thiazolidinedione, and dipeptidyl peptidase-4 inhibitors (49). Drugs, such as pravastatin, rosiglitazone, or coenzyme Q10, improve function and reduce senescence or apoptosis in MSCs (5, 90). Last, both EPCs and MSCs release microparticles, which carry genetic and protein cargo. These noncellular elements are not subject to apoptosis and senescence and might have a longer-lasting impact. Different conditions, disease vs. normal, young vs. aging, affect these paracrine factors from EPCs or MSCs (99). Further studies are needed to determine impact of such cellular or noncellular elements and whether cells derived from CKD patients potentially pack harmful cargo.

Perspectives and Significance

CKD progression to ESRD has become a public health problem. New biomarkers and surrogate end points allow potential intervention and treatment in the earlier stages of CKD, which could reduce or even reverse the progression rate. New omic-based technologies reveal new genomic and epigenomic mechanisms related to CKD incidence and/or progression. AKI has been recognized as a major risk for CKD progression. In addition to optimizing RAAS blockade, new drugs targeting ET-1 and TGF-β, and cell-based regenerative therapy, may further ameliorate the progression of CKD. Last, the gap from suitable experimental models to human disease must be narrowed to be able to efficiently translate new mechanistic understanding to evidence-based treatments for human progressive CKD.

GRANTS

This work was supported in part by National Institute of Diabetes and Digestive and Kidney Diseases Grant DK-56942 (A. B. Fogo).

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

J.Z. and H.Y. drafted manuscript; J.Z., H.Y., and A.B.F. edited and revised manuscript; A.B.F. approved final version of manuscript.

REFERENCES

  • 1.Adler SG, Schwartz S, Williams ME, Arauz-Pacheco C, Bolton WK, Lee T, Li D, Neff TB, Urquilla PR, Sewell KL. Phase 1 study of anti-CTGF monoclonal antibody in patients with diabetes and microalbuminuria. Clin J Am Soc Nephrol 5: 1420–1428, 2010. doi: 10.2215/CJN.09321209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Alchi B, Jayne D, Labopin M, Demin A, Sergeevicheva V, Alexander T, Gualandi F, Gruhn B, Ouyang J, Rzepecki P, Held G, Sampol A, Voswinkel J, Ljungman P, Fassas A, Badoglio M, Saccardi R, Farge D; EBMT Autoimmune Disease Working Party members . Autologous haematopoietic stem cell transplantation for systemic lupus erythematosus: data from the European Group for Blood and Marrow Transplantation registry. Lupus 22: 245–253, 2013. doi: 10.1177/0961203312470729. [DOI] [PubMed] [Google Scholar]
  • 3.Ascon M, Ascon DB, Liu M, Cheadle C, Sarkar C, Racusen L, Hassoun HT, Rabb H. Renal ischemia-reperfusion leads to long term infiltration of activated and effector-memory T lymphocytes. Kidney Int 75: 526–535, 2009. doi: 10.1038/ki.2008.602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Baek SD, Baek CH, Kim JS, Kim SM, Kim JH, Kim SB. Does stage III chronic kidney disease always progress to end-stage renal disease? A ten-year follow-up study. Scand J Urol Nephrol 46: 232–238, 2012. doi: 10.3109/00365599.2011.649045. [DOI] [PubMed] [Google Scholar]
  • 5.Benvenuti S, Cellai I, Luciani P, Deledda C, Baglioni S, Giuliani C, Saccardi R, Mazzanti B, Dal Pozzo S, Mannucci E, Peri A, Serio M. Rosiglitazone stimulates adipogenesis and decreases osteoblastogenesis in human mesenchymal stem cells. J Endocrinol Invest 30: RC26–RC30, 2007. doi: 10.1007/BF03350807. [DOI] [PubMed] [Google Scholar]
  • 6.Bhavsar NA, Köttgen A, Coresh J, Astor BC. Neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule 1 (KIM-1) as predictors of incident CKD stage 3: the Atherosclerosis Risk in Communities (ARIC) Study. Am J Kidney Dis 60: 233–240, 2012. doi: 10.1053/j.ajkd.2012.02.336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Boes E, Fliser D, Ritz E, König P, Lhotta K, Mann JF, Müller GA, Neyer U, Riegel W, Riegler P, Kronenberg F. Apolipoprotein A-IV predicts progression of chronic kidney disease: the mild to moderate kidney disease study. J Am Soc Nephrol 17: 528–536, 2006. doi: 10.1681/ASN.2005070733. [DOI] [PubMed] [Google Scholar]
  • 8.Bolignano D, Lacquaniti A, Coppolino G, Donato V, Campo S, Fazio MR, Nicocia G, Buemi M. Neutrophil gelatinase-associated lipocalin (NGAL) and progression of chronic kidney disease. Clin J Am Soc Nephrol 4: 337–344, 2009. doi: 10.2215/CJN.03530708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Boor P, Ostendorf T, Floege J. Renal fibrosis: novel insights into mechanisms and therapeutic targets. Nat Rev Nephrol 6: 643–656, 2010. doi: 10.1038/nrneph.2010.120. [DOI] [PubMed] [Google Scholar]
  • 10.Brenner BM, Lawler EV, Mackenzie HS. The hyperfiltration theory: a paradigm shift in nephrology. Kidney Int 49: 1774–1777, 1996. doi: 10.1038/ki.1996.265. [DOI] [PubMed] [Google Scholar]
  • 11.Chawla LS, Amdur RL, Amodeo S, Kimmel PL, Palant CE. The severity of acute kidney injury predicts progression to chronic kidney disease. Kidney Int 79: 1361–1369, 2011. doi: 10.1038/ki.2011.42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Chawla LS, Kimmel PL. Acute kidney injury and chronic kidney disease: an integrated clinical syndrome. Kidney Int 82: 516–524, 2012. doi: 10.1038/ki.2012.208. [DOI] [PubMed] [Google Scholar]
  • 13.Chen YT, Cheng BC, Ko SF, Chen CH, Tsai TH, Leu S, Chang HW, Chung SY, Chua S, Yeh KH, Chen YL, Yip HK. Value and level of circulating endothelial progenitor cells, angiogenesis factors and mononuclear cell apoptosis in patients with chronic kidney disease. Clin Exp Nephrol 17: 83–91, 2013. doi: 10.1007/s10157-012-0664-9. [DOI] [PubMed] [Google Scholar]
  • 14.Cho ME, Smith DC, Branton MH, Penzak SR, Kopp JB. Pirfenidone slows renal function decline in patients with focal segmental glomerulosclerosis. Clin J Am Soc Nephrol 2: 906–913, 2007. doi: 10.2215/CJN.01050207. [DOI] [PubMed] [Google Scholar]
  • 15.Choi YJ, Chakraborty S, Nguyen V, Nguyen C, Kim BK, Shim SI, Suki WN, Truong LD. Peritubular capillary loss is associated with chronic tubulointerstitial injury in human kidney: altered expression of vascular endothelial growth factor. Hum Pathol 31: 1491–1497, 2000. doi: 10.1053/hupa.2000.20373. [DOI] [PubMed] [Google Scholar]
  • 16.Coca SG, Singanamala S, Parikh CR. Chronic kidney disease after acute kidney injury: a systematic review and meta-analysis. Kidney Int 81: 442–448, 2012. doi: 10.1038/ki.2011.379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Coresh J, Turin TC, Matsushita K, Sang Y, Ballew SH, Appel LJ, Arima H, Chadban SJ, Cirillo M, Djurdjev O, Green JA, Heine GH, Inker LA, Irie F, Ishani A, Ix JH, Kovesdy CP, Marks A, Ohkubo T, Shalev V, Shankar A, Wen CP, de Jong PE, Iseki K, Stengel B, Gansevoort RT, Levey AS; CKD Prognosis Consortium . Decline in estimated glomerular filtration rate and subsequent risk of end-stage renal disease and mortality. JAMA 311: 2518–2531, 2014. doi: 10.1001/jama.2014.6634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Dagher PC, Mai EM, Hato T, Lee SY, Anderson MD, Karozos SC, Mang HE, Knipe NL, Plotkin Z, Sutton TA. The p53 inhibitor pifithrin-α can stimulate fibrosis in a rat model of ischemic acute kidney injury. Am J Physiol Renal Physiol 302: F284–F291, 2012. doi: 10.1152/ajprenal.00317.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.de Zeeuw D, Akizawa T, Audhya P, Bakris GL, Chin M, Christ-Schmidt H, Goldsberry A, Houser M, Krauth M, Lambers Heerspink HJ, McMurray JJ, Meyer CJ, Parving HH, Remuzzi G, Toto RD, Vaziri ND, Wanner C, Wittes J, Wrolstad D, Chertow GM; BEACON Trial Investigators . Bardoxolone methyl in type 2 diabetes and stage 4 chronic kidney disease. N Engl J Med 369: 2492–2503, 2013. doi: 10.1056/NEJMoa1306033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.de Zeeuw D, Bekker P, Henkel E, Hasslacher C, Gouni-Berthold I, Mehling H, Potarca A, Tesar V, Heerspink HJ, Schall TJ; CCX140-B Diabetic Nephropathy Study Group . The effect of CCR2 inhibitor CCX140-B on residual albuminuria in patients with type 2 diabetes and nephropathy: a randomised trial. Lancet Diabetes Endocrinol 3: 687–696, 2015. doi: 10.1016/S2213-8587(15)00261-2. [DOI] [PubMed] [Google Scholar]
  • 21.de Zeeuw D, Coll B, Andress D, Brennan JJ, Tang H, Houser M, Correa-Rotter R, Kohan D, Lambers Heerspink HJ, Makino H, Perkovic V, Pritchett Y, Remuzzi G, Tobe SW, Toto R, Viberti G, Parving HH. The endothelin antagonist atrasentan lowers residual albuminuria in patients with type 2 diabetic nephropathy. J Am Soc Nephrol 25: 1083–1093, 2014. doi: 10.1681/ASN.2013080830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Dudley AT, Lyons KM, Robertson EJ. A requirement for bone morphogenetic protein-7 during development of the mammalian kidney and eye. Genes Dev 9: 2795–2807, 1995. doi: 10.1101/gad.9.22.2795. [DOI] [PubMed] [Google Scholar]
  • 23.El-Ansary M, Saadi G, Abd El-Hamid SM. Mesenchymal stem cells are a rescue approach for recovery of deteriorating kidney function. Nephrology (Carlton) 17: 650–657, 2012. doi: 10.1111/j.1440-1797.2012.01622.x. [DOI] [PubMed] [Google Scholar]
  • 24.Fang Y, Tian X, Bai S, Fan J, Hou W, Tong H, Li D. Autologous transplantation of adipose-derived mesenchymal stem cells ameliorates streptozotocin-induced diabetic nephropathy in rats by inhibiting oxidative stress, pro-inflammatory cytokines and the p38 MAPK signaling pathway. Int J Mol Med 30: 85–92, 2012. doi: 10.3892/ijmm.2012.977. [DOI] [PubMed] [Google Scholar]
  • 25.Fried LF, Emanuele N, Zhang JH, Brophy M, Conner TA, Duckworth W, Leehey DJ, McCullough PA, O’Connor T, Palevsky PM, Reilly RF, Seliger SL, Warren SR, Watnick S, Peduzzi P, Guarino P; VA NEPHRON-D Investigators . Combined angiotensin inhibition for the treatment of diabetic nephropathy. N Engl J Med 369: 1892–1903, 2013. doi: 10.1056/NEJMoa1303154. [DOI] [PubMed] [Google Scholar]
  • 26.Glassock RJ. Debate: CON position. Should microalbuminuria ever be considered as a renal endpoint in any clinical trial? Am J Nephrol 31: 462–465, 2010. doi: 10.1159/000313553. [DOI] [PubMed] [Google Scholar]
  • 27.Goicoechea M, de Vinuesa SG, Verdalles U, Ruiz-Caro C, Ampuero J, Rincón A, Arroyo D, Luño J. Effect of allopurinol in chronic kidney disease progression and cardiovascular risk. Clin J Am Soc Nephrol 5: 1388–1393, 2010. doi: 10.2215/CJN.01580210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Goldstein SL, Jaber BL, Faubel S, Chawla LS; Acute Kidney Injury Advisory Group of American Society of Nephrology . AKI transition of care: a potential opportunity to detect and prevent CKD. Clin J Am Soc Nephrol 8: 476–483, 2013. doi: 10.2215/CJN.12101112. [DOI] [PubMed] [Google Scholar]
  • 29.Hayek SS, Sever S, Ko YA, Trachtman H, Awad M, Wadhwani S, Altintas MM, Wei C, Hotton AL, French AL, Sperling LS, Lerakis S, Quyyumi AA, Reiser J. Soluble Urokinase Receptor and Chronic Kidney Disease. N Engl J Med 373: 1916–1925, 2015. doi: 10.1056/NEJMoa1506362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Hibbert B, Simard T, Ramirez FD, Pourdjabbar A, Raizman JE, Maze R, Wilson KR, Hawken S, OʼBrien ER. The effect of statins on circulating endothelial progenitor cells in humans: a systematic review. J Cardiovasc Pharmacol 62: 491–496, 2013. doi: 10.1097/FJC.0b013e3182a4027f. [DOI] [PubMed] [Google Scholar]
  • 31.Hosoya T, Kimura K, Itoh S, Inaba M, Uchida S, Tomino Y, Makino H, Matsuo S, Yamamoto T, Ohno I, Shibagaki Y, Iimuro S, Imai N, Kuwabara M, Hayakawa H. The effect of febuxostat to prevent a further reduction in renal function of patients with hyperuricemia who have never had gout and are complicated by chronic kidney disease stage 3: study protocol for a multicenter randomized controlled study. Trials 15: 26, 2014. doi: 10.1186/1745-6215-15-26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Hu B, Gadegbeku C, Lipkowitz MS, Rostand S, Lewis J, Wright JT, Appel LJ, Greene T, Gassman J, Astor BC; African-American Study of Kidney Disease and Hypertension Group . Kidney function can improve in patients with hypertensive CKD. J Am Soc Nephrol 23: 706–713, 2012. doi: 10.1681/ASN.2011050456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Hunsicker LG, Adler S, Caggiula A, England BK, Greene T, Kusek JW, Rogers NL, Teschan PE, Beck G. Predictors of the progression of renal disease in the Modification of Diet in Renal Disease Study. Kidney Int 51: 1908–1919, 1997. doi: 10.1038/ki.1997.260. [DOI] [PubMed] [Google Scholar]
  • 34.Igarashi P, Somlo S. Genetics and pathogenesis of polycystic kidney disease. J Am Soc Nephrol 13: 2384–2398, 2002. doi: 10.1097/01.ASN.0000028643.17901.42. [DOI] [PubMed] [Google Scholar]
  • 35.Inker LA, Astor BC, Fox CH, Isakova T, Lash JP, Peralta CA, Kurella Tamura M, Feldman HI. KDOQI US commentary on the 2012 KDIGO clinical practice guideline for the evaluation and management of CKD. Am J Kidney Dis 63: 713–735, 2014. doi: 10.1053/j.ajkd.2014.01.416. [DOI] [PubMed] [Google Scholar]
  • 36.International HapMap Consortium The International HapMap Project. Nature 426: 789–796, 2003. doi: 10.1038/nature02168. [DOI] [PubMed] [Google Scholar]
  • 37.Jayne D, Tyndall A. Autologous stem cell transplantation for systemic lupus erythematosus. Lupus 13: 359–365, 2004. doi: 10.1191/0961203304lu1027oa. [DOI] [PubMed] [Google Scholar]
  • 38.Jha V, Garcia-Garcia G, Iseki K, Li Z, Naicker S, Plattner B, Saran R, Wang AY, Yang CW. Chronic kidney disease: global dimension and perspectives. Lancet 382: 260–272, 2013. doi: 10.1016/S0140-6736(13)60687-X. [DOI] [PubMed] [Google Scholar]
  • 39.Johnson RJ, Herrera-Acosta J, Schreiner GF, Rodriguez-Iturbe B. Subtle acquired renal injury as a mechanism of salt-sensitive hypertension. N Engl J Med 346: 913–923, 2002. doi: 10.1056/NEJMra011078. [DOI] [PubMed] [Google Scholar]
  • 40.Kao MP, Ang DS, Gandy SJ, Nadir MA, Houston JG, Lang CC, Struthers AD. Allopurinol benefits left ventricular mass and endothelial dysfunction in chronic kidney disease. J Am Soc Nephrol 22: 1382–1389, 2011. doi: 10.1681/ASN.2010111185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Kida Y, Tchao BN, Yamaguchi I. Peritubular capillary rarefaction: a new therapeutic target in chronic kidney disease. Pediatr Nephrol 29: 333–342, 2014. doi: 10.1007/s00467-013-2430-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kohan DE, Barton M. Endothelin and endothelin antagonists in chronic kidney disease. Kidney Int 86: 896–904, 2014. doi: 10.1038/ki.2014.143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Köttgen A, Glazer NL, Dehghan A, Hwang SJ, Katz R, Li M, Yang Q, Gudnason V, Launer LJ, Harris TB, Smith AV, Arking DE, Astor BC, Boerwinkle E, Ehret GB, Ruczinski I, Scharpf RB, Chen YD, de Boer IH, Haritunians T, Lumley T, Sarnak M, Siscovick D, Benjamin EJ, Levy D, Upadhyay A, Aulchenko YS, Hofman A, Rivadeneira F, Uitterlinden AG, van Duijn CM, Chasman DI, Paré G, Ridker PM, Kao WH, Witteman JC, Coresh J, Shlipak MG, Fox CS. Multiple loci associated with indices of renal function and chronic kidney disease. Nat Genet 41: 712–717, 2009. doi: 10.1038/ng.377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Köttgen A, Hwang SJ, Larson MG, Van Eyk JE, Fu Q, Benjamin EJ, Dehghan A, Glazer NL, Kao WH, Harris TB, Gudnason V, Shlipak MG, Yang Q, Coresh J, Levy D, Fox CS. Uromodulin levels associate with a common UMOD variant and risk for incident CKD. J Am Soc Nephrol 21: 337–344, 2010. doi: 10.1681/ASN.2009070725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Köttgen A, Pattaro C, Böger CA, Fuchsberger C, Olden M, Glazer NL, Parsa A, Gao X, Yang Q, Smith AV, O’Connell JR, Li M, Schmidt H, Tanaka T, Isaacs A, Ketkar S, Hwang SJ, Johnson AD, Dehghan A, Teumer A, Paré G, Atkinson EJ, Zeller T, Lohman K, Cornelis MC, Probst-Hensch NM, Kronenberg F, Tönjes A, Hayward C, Aspelund T, Eiriksdottir G, Launer LJ, Harris TB, Rampersaud E, Mitchell BD, Arking DE, Boerwinkle E, Struchalin M, Cavalieri M, Singleton A, Giallauria F, Metter J, de Boer IH, Haritunians T, Lumley T, Siscovick D, Psaty BM, Zillikens MC, Oostra BA, Feitosa M, Province M, de Andrade M, Turner ST, Schillert A, Ziegler A, Wild PS, Schnabel RB, Wilde S, Munzel TF, Leak TS, Illig T, Klopp N, Meisinger C, Wichmann HE, Koenig W, Zgaga L, Zemunik T, Kolcic I, Minelli C, Hu FB, Johansson A, Igl W, Zaboli G, Wild SH, Wright AF, Campbell H, Ellinghaus D, Schreiber S, Aulchenko YS, Felix JF, Rivadeneira F, Uitterlinden AG, Hofman A, Imboden M, Nitsch D, Brandstätter A, Kollerits B, Kedenko L, Mägi R, Stumvoll M, Kovacs P, Boban M, Campbell S, Endlich K, Völzke H, Kroemer HK, Nauck M, Völker U, Polasek O, Vitart V, Badola S, Parker AN, Ridker PM, Kardia SL, Blankenberg S, Liu Y, Curhan GC, Franke A, Rochat T, Paulweber B, Prokopenko I, Wang W, Gudnason V, Shuldiner AR, Coresh J, Schmidt R, Ferrucci L, Shlipak MG, van Duijn CM, Borecki I, Krämer BK, Rudan I, Gyllensten U, Wilson JF, Witteman JC, Pramstaller PP, Rettig R, Hastie N, Chasman DI, Kao WH, Heid IM, Fox CS. New loci associated with kidney function and chronic kidney disease. Nat Genet 42: 376–384, 2010. doi: 10.1038/ng.568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Krenning G, Dankers PY, Drouven JW, Waanders F, Franssen CF, van Luyn MJ, Harmsen MC, Popa ER. Endothelial progenitor cell dysfunction in patients with progressive chronic kidney disease. Am J Physiol Renal Physiol 296: F1314–F1322, 2009. doi: 10.1152/ajprenal.90755.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Lamb EJ, Levey AS, Stevens PE. The Kidney Disease Improving Global Outcomes (KDIGO) guideline update for chronic kidney disease: evolution not revolution. Clin Chem 59: 462–465, 2013. doi: 10.1373/clinchem.2012.184259. [DOI] [PubMed] [Google Scholar]
  • 48.Lambers Heerspink HJ, de Zeeuw D. Debate: PRO position. Should microalbuminuria ever be considered as a renal endpoint in any clinical trial? Am J Nephrol 31: 458–461, 2010. doi: 10.1159/000292501. [DOI] [PubMed] [Google Scholar]
  • 49.Lee PS, Poh KK. Endothelial progenitor cells in cardiovascular diseases. World J Stem Cells 6: 355–366, 2014. doi: 10.4252/wjsc.v6.i3.355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Levey AS, Coresh J. Chronic kidney disease. Lancet 379: 165–180, 2012. doi: 10.1016/S0140-6736(11)60178-5. [DOI] [PubMed] [Google Scholar]
  • 51.Li L, Astor BC, Lewis J, Hu B, Appel LJ, Lipkowitz MS, Toto RD, Wang X, Wright JT Jr, Greene TH. Longitudinal progression trajectory of GFR among patients with CKD. Am J Kidney Dis 59: 504–512, 2012. doi: 10.1053/j.ajkd.2011.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Lipkowitz MS, Freedman BI, Langefeld CD, Comeau ME, Bowden DW, Kao WH, Astor BC, Bottinger EP, Iyengar SK, Klotman PE, Freedman RG, Zhang W, Parekh RS, Choi MJ, Nelson GW, Winkler CA, Kopp JB, SK Investigators . Apolipoprotein L1 gene variants associate with hypertension-attributed nephropathy and the rate of kidney function decline in African Americans. Kidney Int 83: 114–120, 2013. doi: 10.1038/ki.2012.263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Liu N, He S, Ma L, Ponnusamy M, Tang J, Tolbert E, Bayliss G, Zhao TC, Yan H, Zhuang S. Blocking the class I histone deacetylase ameliorates renal fibrosis and inhibits renal fibroblast activation via modulating TGF-beta and EGFR signaling. PLoS One 8: e54001, 2013. doi: 10.1371/journal.pone.0054001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Maahs DM, Caramori L, Cherney DZ, Galecki AT, Gao C, Jalal D, Perkins BA, Pop-Busui R, Rossing P, Mauer M, Doria A, PERL Consortium . Uric acid lowering to prevent kidney function loss in diabetes: the preventing early renal function loss (PERL) allopurinol study. Curr Diab Rep 13: 550–559, 2013. doi: 10.1007/s11892-013-0381-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Macedo E, Mehta RL. Targeting recovery from acute kidney injury: incidence and prevalence of recovery. Nephron Clin Pract 127: 4–9, 2014. doi: 10.1159/000363704. [DOI] [PubMed] [Google Scholar]
  • 56.Mann JF, Green D, Jamerson K, Ruilope LM, Kuranoff SJ, Littke T, Viberti G, ASCEND Study Group . Avosentan for overt diabetic nephropathy. J Am Soc Nephrol 21: 527–535, 2010. doi: 10.1681/ASN.2009060593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Mann JF, Schmieder RE, McQueen M, Dyal L, Schumacher H, Pogue J, Wang X, Maggioni A, Budaj A, Chaithiraphan S, Dickstein K, Keltai M, Metsärinne K, Oto A, Parkhomenko A, Piegas LS, Svendsen TL, Teo KK, Yusuf S, ONTARGET investigators . Renal outcomes with telmisartan, ramipril, or both, in people at high vascular risk (the ONTARGET study): a multicentre, randomised, double-blind, controlled trial. Lancet 372: 547–553, 2008. doi: 10.1016/S0140-6736(08)61236-2. [DOI] [PubMed] [Google Scholar]
  • 58.Mitch WE, Walser M, Buffington GA, Lemann J Jr. A simple method of estimating progression of chronic renal failure. Lancet 2: 1326–1328, 1976. doi: 10.1016/S0140-6736(76)91974-7. [DOI] [PubMed] [Google Scholar]
  • 59.Morrissey J, Hruska K, Guo G, Wang S, Chen Q, Klahr S. Bone morphogenetic protein-7 improves renal fibrosis and accelerates the return of renal function. J Am Soc Nephrol 13, Suppl 1: S14–S21, 2002. [PubMed] [Google Scholar]
  • 60.National Kidney Foundation K/DOQI clinical practice guidelines for chronic kidney disease: evaluation, classification, and stratification. Am J Kidney Dis 39, Suppl 1: S1–S266, 2002. [PubMed] [Google Scholar]
  • 61.Navarro-González JF, Mora-Fernández C, Muros de Fuentes M, Chahin J, Méndez ML, Gallego E, Macía M, del Castillo N, Rivero A, Getino MA, García P, Jarque A, García J. Effect of pentoxifylline on renal function and urinary albumin excretion in patients with diabetic kidney disease: the PREDIAN trial. J Am Soc Nephrol 26: 220–229, 2015. doi: 10.1681/ASN.2014010012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Neuhofer W, Pittrow D. Endothelin receptor selectivity in chronic kidney disease: rationale and review of recent evidence. Eur J Clin Invest 39, Suppl 2: 50–67, 2009. doi: 10.1111/j.1365-2362.2009.02121.x. [DOI] [PubMed] [Google Scholar]
  • 63.Packham DK, Wolfe R, Reutens AT, Berl T, Heerspink HL, Rohde R, Ivory S, Lewis J, Raz I, Wiegmann TB, Chan JC, de Zeeuw D, Lewis EJ, Atkins RC, Collaborative Study Group . Sulodexide fails to demonstrate renoprotection in overt type 2 diabetic nephropathy. J Am Soc Nephrol 23: 123–130, 2012. doi: 10.1681/ASN.2011040378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Papazova DA, Oosterhuis NR, Gremmels H, van Koppen A, Joles JA, Verhaar MC. Cell-based therapies for experimental chronic kidney disease: a systematic review and meta-analysis. Dis Model Mech 8: 281–293, 2015. doi: 10.1242/dmm.017699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Parsa A, Kao WH, Xie D, Astor BC, Li M, Hsu CY, Feldman HI, Parekh RS, Kusek JW, Greene TH, Fink JC, Anderson AH, Choi MJ, Wright JT Jr, Lash JP, Freedman BI, Ojo A, Winkler CA, Raj DS, Kopp JB, He J, Jensvold NG, Tao K, Lipkowitz MS, Appel LJ, AASK Study Investigators, CRIC Study Investigators . APOL1 risk variants, race, and progression of chronic kidney disease. N Engl J Med 369: 2183–2196, 2013. doi: 10.1056/NEJMoa1310345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Parving HH, Brenner BM, McMurray JJ, de Zeeuw D, Haffner SM, Solomon SD, Chaturvedi N, Persson F, Desai AS, Nicolaides M, Richard A, Xiang Z, Brunel P, Pfeffer MA, ALTITUDE Investigators . Cardiorenal end points in a trial of aliskiren for type 2 diabetes. N Engl J Med 367: 2204–2213, 2012. doi: 10.1056/NEJMoa1208799. [DOI] [PubMed] [Google Scholar]
  • 67.Pattaro C, Köttgen A, Teumer A, Garnaas M, Böger CA, Fuchsberger C, Olden M, Chen MH, Tin A, Taliun D, Li M, Gao X, Gorski M, Yang Q, Hundertmark C, Foster MC, O’Seaghdha CM, Glazer N, Isaacs A, Liu CT, Smith AV, O’Connell JR, Struchalin M, Tanaka T, Li G, Johnson AD, Gierman HJ, Feitosa M, Hwang SJ, Atkinson EJ, Lohman K, Cornelis MC, Johansson Å, Tönjes A, Dehghan A, Chouraki V, Holliday EG, Sorice R, Kutalik Z, Lehtimäki T, Esko T, Deshmukh H, Ulivi S, Chu AY, Murgia F, Trompet S, Imboden M, Kollerits B, Pistis G, Harris TB, Launer LJ, Aspelund T, Eiriksdottir G, Mitchell BD, Boerwinkle E, Schmidt H, Cavalieri M, Rao M, Hu FB, Demirkan A, Oostra BA, de Andrade M, Turner ST, Ding J, Andrews JS, Freedman BI, Koenig W, Illig T, Döring A, Wichmann HE, Kolcic I, Zemunik T, Boban M, Minelli C, Wheeler HE, Igl W, Zaboli G, Wild SH, Wright AF, Campbell H, Ellinghaus D, Nöthlings U, Jacobs G, Biffar R, Endlich K, Ernst F, Homuth G, Kroemer HK, Nauck M, Stracke S, Völker U, Völzke H, Kovacs P, Stumvoll M, Mägi R, Hofman A, Uitterlinden AG, Rivadeneira F, Aulchenko YS, Polasek O, Hastie N, Vitart V, Helmer C, Wang JJ, Ruggiero D, Bergmann S, Kähönen M, Viikari J, Nikopensius T, Province M, Ketkar S, Colhoun H, Doney A, Robino A, Giulianini F, Krämer BK, Portas L, Ford I, Buckley BM, Adam M, Thun GA, Paulweber B, Haun M, Sala C, Metzger M, Mitchell P, Ciullo M, Kim SK, Vollenweider P, Raitakari O, Metspalu A, Palmer C, Gasparini P, Pirastu M, Jukema JW, Probst-Hensch NM, Kronenberg F, Toniolo D, Gudnason V, Shuldiner AR, Coresh J, Schmidt R, Ferrucci L, Siscovick DS, van Duijn CM, Borecki I, Kardia SL, Liu Y, Curhan GC, Rudan I, Gyllensten U, Wilson JF, Franke A, Pramstaller PP, Rettig R, Prokopenko I, Witteman JC, Hayward C, Ridker P, Parsa A, Bochud M, Heid IM, Goessling W, Chasman DI, Kao WH, Fox CS, Goessling W, Chasman DI, Kao WH, Fox C. Genome-wide association and functional follow-up reveals new loci for kidney function. PLoS Genet 8: e1002584, 2012. doi: 10.1371/journal.pgen.1002584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Pickering GW, Wright AD, Heptinstall RH. The reversibility of malignant hypertension. Lancet 2: 952–956, 1952. doi: 10.1016/S0140-6736(52)92200-9. [DOI] [PubMed] [Google Scholar]
  • 69.Reinders ME, de Fijter JW, Roelofs H, Bajema IM, de Vries DK, Schaapherder AF, Claas FH, van Miert PP, Roelen DL, van Kooten C, Fibbe WE, Rabelink TJ. Autologous bone marrow-derived mesenchymal stromal cells for the treatment of allograft rejection after renal transplantation: results of a phase I study. Stem Cells Transl Med 2: 107–111, 2013. doi: 10.5966/sctm.2012-0114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Roemeling-van Rhijn M, Reinders ME, de Klein A, Douben H, Korevaar SS, Mensah FK, Dor FJ, IJzermans JN, Betjes MG, Baan CC, Weimar W, Hoogduijn MJ. Mesenchymal stem cells derived from adipose tissue are not affected by renal disease. Kidney Int 82: 748–758, 2012. doi: 10.1038/ki.2012.187. [DOI] [PubMed] [Google Scholar]
  • 71.Ruggenenti P, Cravedi P, Remuzzi G. Mechanisms and treatment of CKD. J Am Soc Nephrol 23: 1917–1928, 2012. doi: 10.1681/ASN.2012040390. [DOI] [PubMed] [Google Scholar]
  • 72.Sabbisetti VS, Waikar SS, Antoine DJ, Smiles A, Wang C, Ravisankar A, Ito K, Sharma S, Ramadesikan S, Lee M, Briskin R, De Jager PL, Ngo TT, Radlinski M, Dear JW, Park KB, Betensky R, Krolewski AS, Bonventre JV. Blood kidney injury molecule-1 is a biomarker of acute and chronic kidney injury and predicts progression to ESRD in type I diabetes. J Am Soc Nephrol 25: 2177–2186, 2014. doi: 10.1681/ASN.2013070758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Schrier RW, Abdallah JG, Weinberger HH, Abraham WT. Therapy of heart failure. Kidney Int 57: 1418–1425, 2000. doi: 10.1046/j.1523-1755.2000.00986.x. [DOI] [PubMed] [Google Scholar]
  • 74.Serrano A, Huang J, Ghossein C, Nishi L, Gangavathi A, Madhan V, Ramadugu P, Ahya SN, Paparello J, Khosla N, Schlueter W, Batlle D. Stabilization of glomerular filtration rate in advanced chronic kidney disease: a two-year follow-up of a cohort of chronic kidney disease patients stages 4 and 5. Adv Chronic Kidney Dis 14: 105–112, 2007. doi: 10.1053/j.ackd.2006.07.009. [DOI] [PubMed] [Google Scholar]
  • 75.Sharma K, Ix JH, Mathew AV, Cho M, Pflueger A, Dunn SR, Francos B, Sharma S, Falkner B, McGowan TA, Donohue M, Ramachandrarao S, Xu R, Fervenza FC, Kopp JB. Pirfenidone for diabetic nephropathy. J Am Soc Nephrol 22: 1144–1151, 2011. doi: 10.1681/ASN.2010101049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Singh N, McNeely J, Parikh S, Bhinder A, Rovin BH, Shidham G. Kidney complications of hematopoietic stem cell transplantation. Am J Kidney Dis 61: 809–821, 2013. doi: 10.1053/j.ajkd.2012.09.020. [DOI] [PubMed] [Google Scholar]
  • 77.Siragy HM, Carey RM. Role of the intrarenal renin-angiotensin-aldosterone system in chronic kidney disease. Am J Nephrol 31: 541–550, 2010. doi: 10.1159/000313363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Smith ER, Lee D, Cai MM, Tomlinson LA, Ford ML, McMahon LP, Holt SG. Urinary neutrophil gelatinase-associated lipocalin may aid prediction of renal decline in patients with non-proteinuric Stages 3 and 4 chronic kidney disease (CKD). Nephrol Dial Transplant 28: 1569–1579, 2013. doi: 10.1093/ndt/gfs586. [DOI] [PubMed] [Google Scholar]
  • 79.Su G, Luan Z, Wu F, Wang X, Tang X, Wu N, Wang K. Long-term follow-up of autologous stem cell transplantation for severe paediatric systemic lupus erythematosus. Clin Rheumatol 32: 1727–1734, 2013. doi: 10.1007/s10067-013-2324-1. [DOI] [PubMed] [Google Scholar]
  • 80.Sugimoto H, Grahovac G, Zeisberg M, Kalluri R. Renal fibrosis and glomerulosclerosis in a new mouse model of diabetic nephropathy and its regression by bone morphogenic protein-7 and advanced glycation end product inhibitors. Diabetes 56: 1825–1833, 2007. doi: 10.2337/db06-1226. [DOI] [PubMed] [Google Scholar]
  • 81.Sun G, Reddy MA, Yuan H, Lanting L, Kato M, Natarajan R. Epigenetic histone methylation modulates fibrotic gene expression. J Am Soc Nephrol 21: 2069–2080, 2010. doi: 10.1681/ASN.2010060633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Tampe B, Tampe D, Zeisberg EM, Müller GA, Bechtel-Walz W, Koziolek M, Kalluri R, Zeisberg M. Induction of Tet3-dependent Epigenetic Remodeling by Low-dose Hydralazine Attenuates Progression of Chronic Kidney Disease. EBioMedicine 2: 19–36, 2015. doi: 10.1016/j.ebiom.2014.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Tampe B, Zeisberg M. Evidence for the involvement of epigenetics in the progression of renal fibrogenesis. Nephrol Dial Transplant 29, Suppl 1: i1–i8, 2014. doi: 10.1093/ndt/gft361. [DOI] [PubMed] [Google Scholar]
  • 84.Tampe D, Zeisberg M. A primer on the epigenetics of kidney fibrosis. Minerva Med 103: 267–278, 2012. [PubMed] [Google Scholar]
  • 85.Tan J, Wu W, Xu X, Liao L, Zheng F, Messinger S, Sun X, Chen J, Yang S, Cai J, Gao X, Pileggi A, Ricordi C. Induction therapy with autologous mesenchymal stem cells in living-related kidney transplants: a randomized controlled trial. JAMA 307: 1169–1177, 2012. doi: 10.1001/jama.2012.316. [DOI] [PubMed] [Google Scholar]
  • 86.Taskapan H, Tam P, Au V, Chow S, Fung J, Nagai G, Roscoe J, Ng P, Sikaneta T, Ting R, Oreopoulos DG. Improvement in eGFR in patients with chronic kidney disease attending a nephrology clinic. Int Urol Nephrol 40: 841–848, 2008. doi: 10.1007/s11255-008-9360-9. [DOI] [PubMed] [Google Scholar]
  • 87.Tuot DS, Plantinga LC, Hsu CY, Jordan R, Burrows NR, Hedgeman E, Yee J, Saran R, Powe NR, Centers for Disease Control Chronic Kidney Disease Surveillance Team . Chronic kidney disease awareness among individuals with clinical markers of kidney dysfunction. Clin J Am Soc Nephrol 6: 1838–1844, 2011. doi: 10.2215/CJN.00730111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Tuttle KR, Bakris GL, Toto RD, McGill JB, Hu K, Anderson PW. The effect of ruboxistaurin on nephropathy in type 2 diabetes. Diabetes Care 28: 2686–2690, 2005. doi: 10.2337/diacare.28.11.2686. [DOI] [PubMed] [Google Scholar]
  • 89.van Koppen A, Joles JA, Bongartz LG, van den Brandt J, Reichardt HM, Goldschmeding R, Nguyen TQ, Verhaar MC. Healthy bone marrow cells reduce progression of kidney failure better than CKD bone marrow cells in rats with established chronic kidney disease. Cell Transplant 21: 2299–2312, 2012. doi: 10.3727/096368912X636795. [DOI] [PubMed] [Google Scholar]
  • 90.van Koppen A, Papazova DA, Oosterhuis NR, Gremmels H, Giles RH, Fledderus JO, Joles JA, Verhaar MC. Ex vivo exposure of bone marrow from chronic kidney disease donor rats to pravastatin limits renal damage in recipient rats with chronic kidney disease. Stem Cell Res Ther 6: 63, 2015. doi: 10.1186/s13287-015-0064-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Venkatachalam MA, Griffin KA, Lan R, Geng H, Saikumar P, Bidani AK. Acute kidney injury: a springboard for progression in chronic kidney disease. Am J Physiol Renal Physiol 298: F1078–F1094, 2010. doi: 10.1152/ajprenal.00017.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Venkatachalam MA, Weinberg JM, Kriz W, Bidani AK. Failed Tubule Recovery, AKI-CKD Transition, and Kidney Disease Progression. J Am Soc Nephrol 26: 1765–1776, 2015. doi: 10.1681/ASN.2015010006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Vilayur E, Harris DC. Emerging therapies for chronic kidney disease: what is their role? Nat Rev Nephrol 5: 375–383, 2009. doi: 10.1038/nrneph.2009.76. [DOI] [PubMed] [Google Scholar]
  • 94.Wang G, Kwan BC, Lai FM, Choi PC, Chow KM, Li PK, Szeto CC. Intrarenal expression of miRNAs in patients with hypertensive nephrosclerosis. Am J Hypertens 23: 78–84, 2010. doi: 10.1038/ajh.2009.208. [DOI] [PubMed] [Google Scholar]
  • 95.Wanner C, Inzucchi SE, Lachin JM, Fitchett D, von Eynatten M, Mattheus M, Johansen OE, Woerle HJ, Broedl UC, Zinman B, EMPA-REG OUTCOME Investigators . Empagliflozin and Progression of Kidney Disease in Type 2 Diabetes. N Engl J Med 375: 323–334, 2016. doi: 10.1056/NEJMoa1515920. [DOI] [PubMed] [Google Scholar]
  • 96.Weis L, Metzger M, Haymann JP, Thervet E, Flamant M, Vrtovsnik F, Gauci C, Houillier P, Froissart M, Letavernier E, Stengel B, Boffa JJ, NephroTest Study Group . Renal function can improve at any stage of chronic kidney disease. PLoS One 8: e81835, 2013. doi: 10.1371/journal.pone.0081835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Wing MR, Devaney JM, Joffe MM, Xie D, Feldman HI, Dominic EA, Guzman NJ, Ramezani A, Susztak K, Herman JG, Cope L, Harmon B, Kwabi-Addo B, Gordish-Dressman H, Go AS, He J, Lash JP, Kusek JW, Raj DS, Chronic Renal Insufficiency Cohort (CRIC) Study . DNA methylation profile associated with rapid decline in kidney function: findings from the CRIC study. Nephrol Dial Transplant 29: 864–872, 2014. doi: 10.1093/ndt/gft537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Yamanaka S, Yokote S, Yamada A, Katsuoka Y, Izuhara L, Shimada Y, Omura N, Okano HJ, Ohki T, Yokoo T. Adipose tissue-derived mesenchymal stem cells in long-term dialysis patients display downregulation of PCAF expression and poor angiogenesis activation. PLoS One 9: e102311, 2014. doi: 10.1371/journal.pone.0102311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Yang HC, Rossini M, Ma LJ, Zuo Y, Ma J, Fogo AB. Cells derived from young bone marrow alleviate renal aging. J Am Soc Nephrol 22: 2028–2036, 2011. doi: 10.1681/ASN.2010090982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Yang L, Besschetnova TY, Brooks CR, Shah JV, Bonventre JV. Epithelial cell cycle arrest in G2/M mediates kidney fibrosis after injury. Nat Med 16: 535–543, 2010. doi: 10.1038/nm.2144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Zeisberg EM, Zeisberg M. The role of promoter hypermethylation in fibroblast activation and fibrogenesis. J Pathol 229: 264–273, 2013. doi: 10.1002/path.4120. [DOI] [PubMed] [Google Scholar]
  • 102.Zeisberg M, Hanai J, Sugimoto H, Mammoto T, Charytan D, Strutz F, Kalluri R. BMP-7 counteracts TGF-beta1-induced epithelial-to-mesenchymal transition and reverses chronic renal injury. Nat Med 9: 964–968, 2003. doi: 10.1038/nm888. [DOI] [PubMed] [Google Scholar]
  • 103.Zhang AH, Tam P, LeBlanc D, Zhong H, Chan CT, Bargman JM, Oreopoulos DG. Natural history of CKD stage 4 and 5 patients following referral to renal management clinic. Int Urol Nephrol 41: 977–982, 2009. doi: 10.1007/s11255-009-9604-3. [DOI] [PubMed] [Google Scholar]
  • 104.Zhang QL, Rothenbacher D. Prevalence of chronic kidney disease in population-based studies: systematic review. BMC Public Health 8: 117, 2008. doi: 10.1186/1471-2458-8-117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Zhu XY, Urbieta-Caceres V, Krier JD, Textor SC, Lerman A, Lerman LO. Mesenchymal stem cells and endothelial progenitor cells decrease renal injury in experimental swine renal artery stenosis through different mechanisms. Stem Cells 31: 117–125, 2013. doi: 10.1002/stem.1263. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Physiology - Renal Physiology are provided here courtesy of American Physiological Society

RESOURCES