Preprint
Review

This version is not peer-reviewed.

Diagnosis and Emerging Biomarkers of Cystic Fibrosis-Related Kidney Disease (CFKD)

A peer-reviewed article of this preprint also exists.

Submitted:

18 June 2025

Posted:

24 June 2025

You are already at the latest version

Abstract
As people with cystic fibrosis (PwCF) live longer, kidney disease is emerging as a sig-nificant comorbidity that increasingly is linked to cardiovascular complications and progression to end-stage kidney disease. In our recent review, we proposed the unifying term CF-related kidney disease (CFKD) to encompass the spectrum of kidney dysfunction observed in this population. Early detection of kidney injury is critical for improving long-term outcomes yet remains challenging due to the limited sensitivity of conventional laboratory tests, particularly in individuals with altered muscle mass and unique CF pathophysiology. Emerging approaches, including novel blood and urinary biomarkers, urinary ex-tra-cellular vesicles, and genetic risk profiling, offer promising avenues for identifying subclinical kidney damage. When integrated with machine learning algorithms, these tools may enable the development of personalized risk stratification models and targeted therapeutic strategies. This precision medicine approach has the potential to transform kidney disease man-agement in PwCF, shifting care from reactive treatment of late-stage disease to proactive monitoring and early intervention.
Keywords: 
;  ;  ;  ;  

1. Introduction

Cystic Fibrosis (CF) is a genetic disorder caused by mutations in the CF Transmembrane Conductance Regulator (CFTR) gene [1]. Since the discovery and clinical use of highly effective modulator therapy (HEMT), which improves mutant CFTR protein expression and/or function, life expectancy for people with CF (PwCF) has increased significantly and resulted in substantial improvements in lung function and quality of life [2,3]. Chronic kidney disease (CKD) has emerged as a notable morbidity in the aging population of PwCF [4,5]. CF-related kidney disease (CFKD) has been proposed as a term for kidney dysfunction affecting PwCF [6,7]. In addition to renal morbidity, CKD is an identified risk factor for cardiovascular events, subsequent end-stage kidney disease (ESKD), and death [8]. The known risk factors for acute kidney injury (AKI) in PwCF include nephrotoxic medications (e.g., aminoglycosides, non-steroidal antiinflammatory drugs), severe pulmonary infections, systemic inflammation, hypoxia, and oxidative stress [9,10,11]. AKI is frequently undetected by conventional assays and may increase the risk of CKD [12]. Evidence-based interventions such as lifestyle modifications and blood pressure control, as well as monitoring medication use during the early stages of kidney injury, may prevent or delay CKD [13,14].
CFTR, abundantly expressed in most segments of the renal tubular epithelium, is implicated in diverse functions that differ from those in the lung [7,15,16,17]. In the proximal tubules of the kidney, CFTR regulates vesicular acidification and the endocytic uptake of low-molecular-weight proteins. In the collecting duct, it plays a role in bicarbonate secretion [reviewed in Hart et al., 2025] [7]. The mechanisms of CFKD are incompletely understood and may include factors associated with CFTR dysfunction in the lung and other organs, including the kidney, cumulative treatment side effects combined with an age-related kidney function decline, as well as genetic factors [7,18,19]. Here, we review conventional and emerging biomarkers for early detection of kidney injury and dysfunction, with a focus on PwCF. To guide future research on CF-specific biomarkers of kidney dysfunction, we describe genetic modifiers associated with worse outcomes in CF and emphasize their potential as CFKD contributors.

2. Conventional Laboratory Tests for Kidney Function and Detection of Kidney Disease

Several laboratory tests performed on blood and urine specimens are typically used in clinical practice to evaluate kidney function. Most tests are not disease-specific and provide only an imperfect estimate of kidney function. Results of several of these tests have to be interpreted together to better understand kidney health and the potential etiology of dysfunction. For this reason, conventional laboratory tests do not meet the criteria of a biomarker, which is defined as a measurable and objectively evaluated characteristic that indicates normal physiological or pathological processes [20].

2.1. Serum Creatinine Concentration and Glomerular Filtration Rate (eGFR)

Serum creatinine concentration and glomerular filtration rate (GFR) are commonly used measurements in clinical practice to estimate kidney function. GFR reflects the kidney’s ability to filter blood, but its direct measurement is often impractical in clinical settings. Serum creatinine concentration is used as a surrogate measure to estimate GFR (eGFR; normal range 90-120 ml/min/1.73m2). Creatinine, a by-product of muscle metabolism, is filtered by the glomeruli. It is not an ideal substance for measuring GFR because it is also secreted by the proximal tubule, leading to an overestimation of GFR by 10-20% [21]. Creatinine levels are influenced by age, sex, muscle mass, nutritional and hydration status, diet, and the use of medications. Its production decreases during sepsis [22].
Serum creatinine is primarily measured using the Jaffe reaction, enzymatic assays, isotope dilution mass spectrometry (IDMS), or high-performance liquid chromatography (HPLC). The Jaffe reaction is a simple and cost-effective colorimetric assay, but it is also limited by susceptibility to interference [23]. Enzymatic methods utilize specific enzymes, providing greater specificity and less interference than the Jaffe method, although they are more expensive [24]. IDMS is considered the gold standard for creatinine measurement, providing high precision and serving as a reference method for standardization, but its high cost and limited availability restrict its routine clinical use [25]. HPLC separates creatinine from other serum components before detection, minimizing interference. While it is highly specific and accurate, it is time-consuming and requires specialized equipment [26]. GFR estimation methods rely on the assumption of a steady state—a condition in which the rate of creatinine production by the body equals the rate of its clearance by the kidneys. Creatinine reflects eGFR during the steady state, and the utility of serum creatinine declines as kidney disease progresses [27]. In response to injury, the kidney deploys compensatory mechanisms, leading to hyperfiltration through functioning glomeruli that maintain normal or even elevated eGFR, thereby masking the progressive loss of nephrons. Serum creatinine levels only begin to increase after significant kidney damage has occurred, with renal function declining by as much as 50% before noticeable changes in creatinine are detected [28]. Therefore, the correlation between kidney function and eGFR is non-linear; eGFR does not accurately predict early kidney disease or define the mechanisms of injury [29]. An overestimation of eGFR due to creatinine secretion by the kidney proximal tubule further complicates measurements.

2.2. Serum Cystatin C

Cystatin C, a substance released by all nucleated cells, has been used to measure eGFR (recommendations from the National Kidney Foundation & American Society of Nephrology NKF-ASN Task Force) [30]. It is particularly useful when combined with serum creatinine measurements [31]. Various eGFR formulas exist, based on measurements of serum creatinine and/or Cystatin C, age groups, sex, and population characteristics. Some of the most commonly used formulas include those from the Chronic Kidney Disease in Children (CKiD) Study and the CKD-EPI, which are tailored to specific groups for more accurate estimation of kidney function. For example, the CKiD U25 eGFR formula is specifically designed for children and young adults aged 1-25 years. Other formulas are optimized for specific populations, including those with CKD or older adults (Table 1) [32,33,34,35,36]. Two independent studies concluded that cystatin C clearance outperforms eGFR in PwCF [37,38]. However, none of the current formulas for eGFR are validated in PwCF, and thus the correlation with GFR in PwCF is unclear [39].

2.3. Urine Protein Composition and Concentration

Urine protein composition and concentration are used to assess glomerular and tubular integrity. Urinary albumin excretion is typically classified into three categories: normoalbuminuria (<30 mg/day or UACR <30 mg/g), microalbuminuria (30–300 mg/day or UACR 30–300 mg/g), and macroalbuminuria (>300 mg/day or UACR >300 mg/g) [40]. Excessive urinary albumin concentration (macroalbuminuria) indicates glomerular damage. By contrast, tubular damage is suggested by elevated concentrations of urinary low molecular weight (LMW; molecular weight <40kDa) proteins, such as beta-2 microglobulin (β2MG) and retinol-binding protein (RBP) in the presence of normoalbuminuria [41]. Urinary LMW proteins are typically measured using various biochemical and electrophoretic techniques, including enzyme-linked immunosorbent assay (ELISA), sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), liquid chromatography-tandem mass spectrometry (LC-MS), and Urine Protein Electrophoresis (UPEP), which is a clinical laboratory test [42,43,44].
Microalbuminuria can be at least partially mitigated by tubular reabsorption of albumin. This process exerts metabolic stress on the renal proximal tubule where the reabsorption occurs, leading to tubular damage, tubulointerstitial fibrosis, and CKD progression over months to years. For all these reasons, worsening proteinuria is a test of established kidney injury and its progression.
According to the Kidney Disease Improving Global Outcomes (KDIGO) guidelines, AKI is defined by an increase in serum creatinine (within 2–7 days) and/or decrease in urinary output, while CKD is determined based on decreased eGFR <60 mL/min/1.73m2 and albuminuria persistent for more than 3 months [14]. The Fractional Excretion of Sodium (FeNa) and Fractional Excretion of Urea (FeUrea) are key tests for distinguishing prerenal from intrinsic renal AKI [45]. FeNa < 1% suggests prerenal AKI, while FeNa > 2% indicates intrinsic renal AKI. FeUrea < 35% suggests prerenal AKI, and FeUrea > 50% suggests intrinsic AKI, especially in patients on diuretics. Urine specific gravity and osmolality also aid in the diagnosis; high specific gravity (>1.020) and osmolality (>500 mOsm/kg) point to prerenal AKI, while low specific gravity (<1.010) and osmolality (<300 mOsm/kg) suggest intrinsic renal injury, such as acute tubular necrosis (ATN) [46].

2.4. Other Tests

Several other tests can provide valuable insight into kidney health. Urinalysis is a standard diagnostic tool used to evaluate kidney function and detect abnormalities in urine composition. The presence of red blood cells (RBCs) in urine (hematuria) can indicate glomerular disease or lower urinary tract abnormalities such as nephrolithiasis. Pyuria (the presence of white blood cells, WBCs) suggests urinary tract infection or interstitial nephritis. Cellular casts, including RBC casts, indicate glomerulonephritis, while WBC casts suggest interstitial nephritis or infection. Granular casts are frequently observed in ATN and indicate tubular injury, while hyaline casts, which can be found in concentrated urine or with dehydration, are less specific. Crystals in the urine can indicate metabolic disturbances and nephrolithiasis [47]. Kidney biopsy and histological evaluation of kidney tissue provide the most specific diagnosis and may predict the severity and prognosis of kidney disease. The kidney complications in PwCF have been recently reviewed [7]. Many of the standard clinical tests reviewed above are used for their diagnosis (Table 2) [7,48,49,50,51,52,53,54,55,56,57,58,59,60].

3. Emerging Biomarkers of Early Kidney Injury in the General Population and PwCF

As described above, conventional tests report already-established kidney damage and are insensitive to subclinical or early injury. In contrast, novel biomarkers in the blood and urine may provide insights into the timing, severity, and site of kidney injury [61]. Molecular alterations revealed in genomic, proteomic, and metabolomic studies may recognize renal damage in the preclinical phase and provide insight into the pathophysiology of early kidney disease. Urinary biomarkers have already been recommended for detecting drug-induced tubular injury in early clinical trials [62].
Kidney injury molecule (KIM)-1 showed a strong negative correlation with eGFR in both AKI and CKD settings [63,64,65]. KIM-1 is a phosphatidylserine receptor expressed on the proximal tubules and targets apoptotic cells to lysosomes. Interleukin-18 (IL-18) and neutrophil gelatinase-associated lipocalin (NGAL) are also considered potential biomarkers for kidney injury [66,67,68]. The cell cycle arrest biomarkers tissue inhibitor of metalloproteinases-2 (TIMP-2) and insulin-like growth factor-binding protein 7 (IGFBP7) have emerged as promising indicators for detecting acute kidney injury (AKI), particularly in critically ill patients and those undergoing cardiac surgery. Recognizing its diagnostic potential, the NephroCheck™ test, which quantifies the product of [TIMP-2] × [IGFBP7], was approved by the U.S. Food and Drug Administration (FDA) in 2014 for clinical use in intensive care unit (ICU) settings to assess the risk of developing moderate to severe AKI [69]. Selenium-binding protein 1 (SBP1) plays a multifaceted role in cellular processes, exhibiting predominant expression in proximal tubular cells under normal conditions. Elevated urinary SBP1 levels have been identified as an early and sensitive biomarker of AKI, outperforming traditional markers like NGAL and TIMP-2 [70, 71. Kidney injury and inflammatory markers and tests across CKD stages are summarized in Table 3 [70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85].
Even in the absence of albuminuria and with normal eGFR, PwCF exhibit evidence of both tubular cell dysfunction and renal injury, including decreased levels of urinary EGF, and elevated levels of urinary tubular injury markers such as KIM-1, TFF3, β2MG, Cystatin C, and N-acetyl-β-D-glucosaminidase (NAG) [68]. These findings confirm that renal tubular injury occurs before significant glomerular dysfunction becomes evident, and decreased uEGF may suggest early signs of CKD in some PwCF [68,86,87]. Levels of KIM-1 and β2MG are also increased in PwCF during pulmonary exacerbations and correlate with lung function decline, pointing to the role of lung infection and inflammation in contributing to renal injury [68]. Increased urinary NAG levels suggest subclinical tubular kidney injury in PwCF undergoing aminoglycoside therapy, including nebulized tobramycin in children [88,89]. Elevated urinary soluble Fas (sFas) concentrations during aminoglycoside treatment are associated with the development of CKD in PwCF [90]. While these observations were limited to a few tubular injury markers, which have yet to be systematically studied in PwCF, they support the concept of a novel tool for the rapid and early detection of kidney injury before elevation of serum creatinine or urinary protein.
Extracellular vesicles (EVs) are a source of emerging site- and disease-specific kidney injury markers [91]. EVs are membrane-bound nanosized particles released by cells that play a significant role in intercellular communication by transferring signals capable of altering target cell function and influencing the pathophysiology of various diseases, including CF [92]. Urinary extracellular vesicles (uEVs), which are EVs present in urine, have been shown to mediate the crosstalk between different cell populations during nephrogenesis, amplification of kidney injury, inflammation, fibrosis, and regeneration [93]. Intercellular communication in the kidney occurs not only between mesangial, endothelial, and podocyte cells within the glomeruli, but also between glomerular and tubular compartments and among different tubular cell types [94].
Both the number of uEVs released and their contents (proteins, lipids, DNA, and RNA species) change under cellular stress and have been investigated as biomarkers for kidney disease [95]. It has been proposed that the characteristic signatures of the EV cargo may be leveraged as markers for location-specific kidney disease [96,97,98]. Because AKI frequently involves acute tubular damage that can progress to chronic injury characterized by interstitial fibrosis, the crosstalk between the tubular and interstitial compartments has become a key focus for uncovering new mechanisms underlying maladaptive repair processes and the progression from AKI to CKD [94]. A panel of proteins and micro(mi)RNAs have been associated explicitly with AKI versus CKD, glomerular versus tubular renal injury, specific kidney disease processes, and maladaptation after AKI [93]. Thus, elevated levels of specific EV markers may provide mechanistic information, and an early index of AKI may allow for monitoring of CKD progression in PwCF. However, further research is needed to validate these markers in PwCF and to explore their application in early diagnosis and monitoring of kidney disease progression.
Altered levels of specific proteins in uEVs have been linked to kidney injury. NGAL in uEVs, but not in free urine, rises in delayed graft function post-transplant, indicating EVs may protect and enrich certain biomarkers [99]. Panich et al. identified exosomal activating transcription factor-3 (ATF3) as an early marker of sepsis-induced AKI [100]. Increased aquaporin-2 (AQP2) in uEVs signals collecting duct injury after transplantation, while low preoperative podocalyxin levels suggest early podocyte damage and risk of postoperative AKI [101,102]. Decreased CD133 (Prominin-1), a renal progenitor cell marker, in uEVs is seen in both AKI and CKD, reflecting reduced regenerative capacity [103]. MicroRNAs such as miR-125a-5p and miR-10a-5p are also reduced in uEVs of patients who later develop severe AKI, highlighting their potential in risk stratification [101]. Classical exosomal markers CD9 and CD63 are often elevated in uEVs of kidney transplant recipients with AKI, indicating increased vesicular trafficking during injury [102]. Wilms tumor-1 (WT-1), essential for podocyte integrity, is detected in uEVs as a marker of glomerular damage in various kidney diseases [104]. In chronic kidney injury, p16INK4a, a cellular senescence marker, is found in uEVs and linked to hypertensive nephropathy, providing insights into renal aging and fibrosis [105]. Takizawa et al. used a Tim4-based ELISA to measure uEV markers, including distal tubule/collecting duct-specific Mucin 1 (MUC1) and proximal tubule-specific maltase-glucoamylase (MGAM), showing the MGAM/MUC1 ratio rises as kidney function declines in CKD patients [106]. Together, these uEV-associated markers provide valuable insights into molecular events in AKI and CKD and show promise for precision nephrology. However, they have not yet been validated in PwCF.

4. Potential Genetic Modifiers of CFKD in PwCF

CKD and CF share an extensive literature regarding genetic modifiers of disease progression, with potential overlapping mechanisms. Although the genetic modifiers influencing disease severity have been extensively studied and separately validated in CF and CKD, it remains unknown how the modifiers associated with CF lung disease or CF-related diabetes affect the risk of CF-related kidney disease [107,108,109,110]. It is also unclear whether the risks of CKD progression identified in the general population have a similar effect in PwCF. Below, we review several gene polymorphisms associated with a potential for influencing the risk and severity of CKD in PwCF (Table 4) [111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166].
Transforming growth factor β1 (TGFβ1) is an established genetic modifier in CF. Several TGFβ1 polymorphisms are associated with CF progression and P. aeruginosa infection [164]. Elevated TGFβ1 levels downregulate CFTR mRNA levels through miR-145-5p [167]. Besides CFTR, TGFβ1 also regulates Calcium-activated Chloride Conductance (CaCC), thereby maintaining normal hydration of epithelial surfaces, including the airways and colon [168]. Aberrant TGFβ1 signaling drives HNF1B-Related Autosomal Dominant Tubulointerstitial Kidney Disease characterized by tubular cysts, renal fibrosis, and progressive decline in kidney function [150]. Genetic variations in TGFβ1 are also associated with susceptibility to IgA nephropathy [138], autosomal dominant polycystic kidney disease (ADPKD) [156], and CKD [160]. Furthermore, IgA nephropathy is the most common glomerular disease as one of the kidney complications in PwCF [7].
Tumor necrosis factor α (TNFα) (-308 GA, rs1800629) polymorphism is associated with CF in diverse populations [143]. The TNFα +691g ins/del polymorphic locus is associated with the severity of CF lung disease and the age of onset of P. aeruginosa infection [132]. Different studies have found TNFα-308GA promoter polymorphism (rs1800629) that was associated with high TNFα transcription, CF, and AKI severity [116,127,142]. High levels of TNFα induce scaffold protein FIP2, which disrupts the localization of polycystin 2 (PC2) to the plasma membrane and primary cilia in ADPKD [134,135].
Angiotensin-converting enzyme (ACE) is an enzyme in the renin-angiotensin-aldosterone system that is essential for regulating blood pressure and fluid balance. ACE gene D/I polymorphism is a modulator of the severity of CF [140]. ACE2 expression and localization are regulated by the CFTR gene, suggesting a possible role in progression of CF disease [161,166]. ACE also regulates kidney function, and ACE inhibitors are commonly used to treat kidney disease [169]. Among hypertensive patients, the ACE-DD genotype has been shown to be a risk factor for the causation and development of chronic kidney failure [147]. In severe forms of IgA nephropathy (IgAN), ID/DD genotypes are associated with chronic lesions, such as capsular adhesions or glomerulosclerosis and proteinuria [115].
Mannose-binding lectin (MBL2), a member of C-type lectin family activates the complement system during inflammation and can cause both pathogen clearance and tissue injury [146]. In CF, low MBL2 levels are associated with decreased survival and increased susceptibility to P. aeruginosa infection and worse lung function [117,154]. A nonlinear association between MBL levels and renal outcome has been found in IgAN, with both MBL deficiency and excess independently linked to poor renal outcomes, suggesting that MBL contributes to IgAN progression through multiple mechanisms [149,165]. High serum MBL levels are also associated with the development and progression of diabetic nephropathy [155].
α1-antitrypsin (AAT) is the most abundant proteinase inhibitor within the lung and prevents tissue damage caused by inflammation. AAT is considered a genetic modifier in CF, and exogenous AAT has been proposed as a potential therapy for CF [137,145]. However, two separate clinical studies suggest a protective effect of low to moderate levels of AAT on the progression of CF [119,123]. There are three alleles for AAT: M (normal), S (264Glu→Val), and Z (342Glu→Lys). The prevalence of S and Z alleles is approximately 12% in the CF population. Homozygous S and Z alleles result in 60% and 10% of plasma AAT levels in the homozygous state when compared with the homozygous M allele [114]. In CKD, AAT has a protective effect [159]. Two studies showed contradictory results regarding the effect of S and Z alleles on the age of onset of chronic P. aeruginosa acquisition in patients with CF [112,122]. Another polymorphism in the 3’UTR of AAT (G1237A) is associated with a small rise in AAT levels during the acute inflammatory conditions in CF [114], although a later study with a large sample size found no association between G1237A and lung functions in CF patients [119]. A rapid rise in AAT levels can predict AKI in experimental and clinical settings [144,153]. S and Z alleles have been associated with high levels of the antigen of antineutrophil cytoplasm antibodies (ANCA) in Granulomatosis with polyangiitis [111]. In conclusion, despite the considerable amount of data, the impact of AAT on CF phenotype is still unclear, and its role in CF and CKD warrants further investigations.
It has been shown that stimulation of β-adrenoceptor (β-AR) results in increased alveolar fluid clearance and ciliary beat frequency in PwCF [120]. The Gly16Glu27 β2AR genetic variant upregulates CFTR activity in adult CF patients [133]. A similar effect is observed with β2AR agonist salbutamol and ritodrine, suggesting that the effect increases β2AR activity [148]. In the kidney, β2AR is expressed in proximal tubules, glomeruli, and podocytes [152]. Similarly to its role in CF, β2AR agonist reduces the proinflammatory response to renal inflammation [157].
Interleukin-10 (IL-10) is an anti-inflammatory cytokine present in low levels in CF patients. In a study of 220 CF patients, IL-10 haplotype GCC/ACC was significantly associated with P. Aeruginosa infection and CF severity [136]. In another study of 378 patients with CF, a significant association was found between the −1082GG genotype and colonization with A. fumigatus and allergic bronchopulmonary aspergillosis [131]. IL-10 plays an important role in renal physiology, AKI, and progression of chronic renal failure [141]. Polymorphism in IL-10 is associated with AKI (AKI) [127]. IL-10 -1082 A/G polymorphism was associated with increased risks of AKI [158] and primary glomerulonephritis [125].
Nitric oxide synthase (NOS) is an immune modulator and vasodilator. PwCF typically show below normal levels of exhaled NO [129,130]. NOS1 and NOS2 polymorphisms are associated with disease severity and inflammation. More specifically, the number of AAT repeats in the NOS1 gene is negatively correlated with nasal and expired NO in PwCF, and positively correlated with P. aeruginosa and A. fumigatus infection [118,121]. Separate studies have found that the number of GT repeats in NOS1 promoter and G847T polymorphism in the NOS3 gene are positively associated with increased NO production and slower decline in lung function [124,128]. NO levels are reduced in CKD [170]. Experimental NOS inhibition is associated with systemic and glomerular hypertension, glomerular ischemia, glomerulosclerosis, tubulointerstitial injury, and proteinuria [139]. Meta-analysis of 13 studies suggests the presence of the two NOS3 gene polymorphisms, Glu298Asp polymorphisms 4 b/a and -786T>C that have been associated with an increased risk of ESKD in patients with CKD and ADPKD [151,163].
Glutathione S-transferase (GST) is an antioxidant enzyme that conjugates hydroperoxides with glutathione, thereby mitigating tissue damage. CF patients homozygous for glutathione S-transferase M1 (GSTM1) allele have worse chest radiographic scores and worse Shwachman-Kulczycki (SK) scores of CF disease severity [113]. Another polymorphism of the GSTM3*B allele contributes to clinical severity in CF [126]. A meta-analysis found an association between GSTM1, GSTT1, and GSTP1 genetic polymorphisms and the risk of ESKD [146]. Another study of 674 children identified the association of GSTM1 deletion with more rapid progression of pediatric CKD [162]. In conclusion, GWAS studies are needed to examine the role of specific CF genetic modifiers in CFKD risk.
ß-catenin has emerged as a key contributor to kidney fibrosis in the context of CFTR dysfunction. Zhang et al. showed a clear upregulation of the ß-catenin pathway in renal epithelial cells with CFTR knockdown and in the kidneys of F508del mice subjected to unilateral ureteral obstruction (UUO), a well-established model of kidney fibrosis [17]. Furthermore, knockdown of CFTR reduced the expression of tight junction proteins Occludin and ZO-1 in kidney tubular cells, reasoning that it could be a cause of increased leakage of low molecular weight proteins as seen in the urine of PwCF. This, along with enhanced epithelial-to-mesenchymal and fibrosis seen in CFTR mutant mice, may underlie the frequent kidney disease observed in CF.

5. Future Directions of Research on Kidney Function in PwCF

Elucidating the mechanisms of kidney injury is essential for its early detection and prevention. A critical question in CFKD is to what extent the increased prevalence of AKI and CKD results from extrarenal causes, including CF lung disease and nephrotoxic drug exposure, as opposed to intrinsic susceptibility to tubulointerstitial or glomerular injury resulting from renal CFTR dysfunction, decreased nephron endowment [171], pH sensitivity, or pro-fibrotic signaling. Although many of these pathways have been observed in human biospecimens, separating a primary defect from the response to injury is difficult. The recent development of excellent CF animal models [172,173,174], may allow for in vivo distinction of the effect of CFTR dysfunction on kidney health, and the impact of CFKD on the aging population of PwCF in the era of HEMT. These models are increasingly responsive to clinically relevant CFTR modulators [175,176], helping to distinguish the response to nephrotoxic injury on or off therapy. For example, the humanized G551D CF rat model may prove a valuable tool for studying CF kidney disease, as it spontaneously develops lung disease [177], responds to the CFTR modulator Ivacaftor [176], and offers an appealing husbandry profile for reproduction and cost. Similarly, CFTR-knockout ferrets, which mimic human lung and pancreatic disease, offer a promising model to study CFTR-related kidney damage [178]. Additionally, machine learning presents a promising approach with the potential to identify PwCF at risk for kidney disease. Machine learning algorithms are capable of predicting the onset of CKD in asymptomatic individuals, improving risk stratification for progression or complications, and identifying distinct kidney phenotypes or subtypes by linking clinical features to underlying pathological mechanisms [179].

Author Contributions

Conceptualization, H.Y. and A.S.-U.; methodology, H.Y., A.S.-U., S.S.-G., W.T.H.; investigation, H.Y. and A.S.-U.; writing—original draft preparation, H.Y. and A.S.-U.; writing—review and editing, H.Y., M.K., H.B.G., U.E., W.T.H., S.S.-G., M.G., A.S.-U.; supervision, A.S.-U. All authors have read and agreed to the published version of the manuscript.

Funding

This manuscript was funded by the Cystic Fibrosis Foundation grant, CFF SWIATE24A0-MPI (to A.S.-U), SKOPEL24A0-MPI (to S.S.-G), and HARRIS24A0-MPI (to W.T.H.); National Institutes of Health grants R01HL144539, and P50DK096373-11 (to A.S.-U.); the American Diabetes Association grant #7-22-ICTSPM-19/GR100086 (to U.E.).

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
CF Cystic Fibrosis
CFTR CF Transmembrane Conductance Regulator
HEMT Highly effective modulator therapy
PwCF People with CF
CKD Chronic kidney disease
CFKD CF-related kidney disease
ESKD End-stage kidney disease
AKI Acute kidney injury
GFR Glomerular filtration rate
eGFR Estimate GFR
IDMS Isotope dilution mass spectrometry
HPLC High-performance liquid chromatography
CKiD Chronic Kidney Disease in Children
CKD-EPI Chronic kidney disease epidemiology collaboration
MDRD Modification of diet in renal disease
UACR Urine albumin to creatinine ratio
LMW Low molecular weight
β2MG Beta-2 microglobulin
RBP Retinol-binding protein
ELISA Enzyme-linked immunosorbent assay
SDS-PAGE Sodium dodecyl sulfate-polyacrylamide gel electrophoresis
LC-MS Liquid chromatography-tandem mass spectrometry
UPEP Urine protein electrophoresis
KDIGO Kidney Disease Improving Global Outcomes
FeNa Fractional excretion of sodium
FeUrea Fractional excretion of urea
ATN Acute tubular necrosis
RBC Red blood cell
WBC White blood cell
BUN Blood urea nitrogen
SAP Serum amyloid P component
IgA Immunoglobulin A
UPCR Urine protein to creatinine ratio
UCaCR Urine calcium to creatinine ratio
KIM-1 Kidney injury molecule-1
IL-18 Interleukin-18
TIMP-2 Tissue inhibitor of metalloproteinases-2
IGFBP7 Insulin-like growth factor-binding protein 7
FDA Food and Drug Administration
SBP1 Selenium-binding protein 1
NGAL Neutrophil gelatinase-associated lipocalin
TNFα Tumor necrosis factor α
NAG N-acetyl-β-D-glucosaminidase
sFas Soluble Fas
EVs Extracellular vesicles
uEVs Urinary extracellular vesicles
ATF3 Activating transcription factor-3
AQP2 Aquaporin-2
CD133 Prominin-1
WT-1 Wilms tumor-1
MUC1 Mucin 1
MGAM Maltase-glucoamylase
TGFβ1 Transforming growth factor β1
HNF1β Hepatocyte nuclear factor 1-beta
ADTKD Autosomal dominant tubulointerstitial kidney disease
IgAN Immunoglobulin A nephropathy
ACE Angiotensin-converting enzyme
MBL2 Mannose-binding lectin
AAT α1-antitrypsin
ANCA Antineutrophil cytoplasm antibodies
β2AR Beta-2-adrenergic receptor
PC2 Polycystin 2
ADPKD Autosomal dominant polycystic kidney disease
IL-10 Interleukin-1
NOS Nitric oxide synthase
NO Nitric oxide
GST Glutathione S-transferase
CaCC Calcium-activated chloride conductance
GSTM1 Glutathione S-transferase M1
SK Shwachman-Kulczycki

References

  1. Chen Q, Shen Y, Zheng J. A review of cystic fibrosis: Basic and clinical aspects. Animal Model Exp Med. 2021 Sep;4(3):220-32.
  2. Bell SC, Mall MA, Gutierrez H, Macek M, Madge S, Davies JC, et al. The future of cystic fibrosis care: a global perspective. Lancet Respir Med. 2020 Jan;8(1):65-124.
  3. Martin C, Burnet E, Ronayette-Preira A, de Carli P, Martin J, Delmas L, et al. Patient perspectives following initiation of elexacaftor-tezacaftor-ivacaftor in people with cystic fibrosis and advanced lung disease. Respir Med Res. 2021 Nov;80:100829.
  4. Schechter MS, Stecenko AA. Chronic kidney disease: a new morbidity of cystic fibrosis or an old morbidity of diabetes mellitus? Am J Respir Crit Care Med. United States: 2011. Vol. 10, p. 1101-2.
  5. Stevanovic M, Graber ML. Characteristics of US Individuals With Cystic Fibrosis and ESRD: SA-PO324. Journal of the American Society of Nephrology. 2022;33(11S):694.
  6. Nowakowski ACH. Cystic Fibrosis Kidney Disease: 10 Tips for Clinicians. Front Med (Lausanne). 2018;5:242.
  7. Hart M, Kumar M, Goswami HB, Harris WT, Skopelja-Gardner S, Swiatecka-Urban A. Cystic fibrosis-related kidney disease-emerging morbidity and disease modifier. Pediatr Nephrol. 2025 Mar 17.
  8. Jankowski J, Floege J, Fliser D, Böhm M, Marx N. Cardiovascular Disease in Chronic Kidney Disease: Pathophysiological Insights and Therapeutic Options. Circulation. 2021 Mar 16;143(11):1157-72.
  9. Fu Q, Colgan SP, Shelley CS. Hypoxia: The Force that Drives Chronic Kidney Disease. Clin Med Res. 2016 Mar;14(1):15-39.
  10. Muirhead C, Lim JY, Lapidus J, MacDonald K. Evaluation of the Risk for Acute Kidney Injury in Adult Cystic Fibrosis Patients Receiving Concomitant Vancomycin and Tobramycin. Cureus. 2017 Dec 6;9(12):e1912.
  11. Mendes RS, Silva PL, Robba C, Battaglini D, Lopes-Pacheco M, Caruso-Neves C, et al. Advancements in understanding the mechanisms of lung-kidney crosstalk. Intensive Care Med Exp. 2024 Sep 16;12(1):81.
  12. Leung KC, Tonelli M, James MT. Chronic kidney disease following acute kidney injury-risk and outcomes. Nat Rev Nephrol. 2013 Feb;9(2):77-85.
  13. KDIGO 2021 Clinical Practice Guideline for the Management of Blood Pressure in Chronic Kidney Disease. Kidney Int. 2021 Mar;99(3s):S1-s87.
  14. KDIGO 2024 Clinical Practice Guideline for the Evaluation and Management of Chronic Kidney Disease. Kidney Int. 2024 Apr;105(4s):S117-s314.
  15. Crawford I, Maloney PC, Zeitlin PL, Guggino WB, Hyde SC, Turley H, et al. Immunocytochemical localization of the cystic fibrosis gene product CFTR. Proc Natl Acad Sci U S A. 1991 Oct 15;88(20):9262-6.
  16. Morales MM, Carroll TP, Morita T, Schwiebert EM, Devuyst O, Wilson PD, et al. Both the wild type and a functional isoform of CFTR are expressed in kidney. Am J Physiol. 1996 Jun;270(6 Pt 2):F1038-48.
  17. Zhang JT, Wang Y, Chen JJ, Zhang XH, Dong JD, Tsang LL, et al. Defective CFTR leads to aberrant β-catenin activation and kidney fibrosis. Sci Rep. 2017 Jul 12;7(1):5233.
  18. Quon BS, Mayer-Hamblett N, Aitken ML, Smyth AR, Goss CH. Risk factors for chronic kidney disease in adults with cystic fibrosis. Am J Respir Crit Care Med. 2011 Nov 15;184(10):1147-52.
  19. Berg KH, Ryom L, Faurholt-Jepsen D, Pressler T, Katzenstein TL. Prevalence and characteristics of chronic kidney disease among Danish adults with cystic fibrosis. J Cyst Fibros. 2018 Jul;17(4):478-83.
  20. Biomarkers Definitions Working, G. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther. 2001 Mar;69(3):89-95.
  21. Matthew DBaZQ. Better nephrology for mice—and man. Kidney International. 2010;77(6):487-89.
  22. Doi K, Yuen PS, Eisner C, Hu X, Leelahavanichkul A, Schnermann J, et al. Reduced production of creatinine limits its use as marker of kidney injury in sepsis. J Am Soc Nephrol. 2009 Jun;20(6):1217-21.
  23. Toora BD, Rajagopal G. Measurement of creatinine by Jaffe's reaction--determination of concentration of sodium hydroxide required for maximum color development in standard, urine and protein free filtrate of serum. Indian J Exp Biol. 2002 Mar;40(3):352-4.
  24. Moss GA, Bondar RJ, Buzzelli DM. Kinetic enzymatic method for determining serum creatinine. Clin Chem. 1975 Sep;21(10):1422-6.
  25. Miller WG, Myers GL, Ashwood ER, Killeen AA, Wang E, Thienpont LM, et al. Creatinine measurement: state of the art in accuracy and interlaboratory harmonization. Arch Pathol Lab Med. 2005 Mar;129(3):297-304.
  26. Yuen PS, Dunn SR, Miyaji T, Yasuda H, Sharma K, Star RA. A simplified method for HPLC determination of creatinine in mouse serum. Am J Physiol Renal Physiol. 2004 Jun;286(6):F1116-9.
  27. Tian Q, Wu W, Liu J, Wu Z, Yao W, Ding J, et al. Dimensional heterostructures of 1D CdS/2D ZnIn(2)S(4) composited with 2D graphene: designed synthesis and superior photocatalytic performance. Dalton Trans. 2017 Feb 28;46(9):2770-77.
  28. Gounden V, Bhatt H, Jialal I. Renal Function Tests. [Updated 2024 Jul 27]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2025 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK507821/.
  29. Mizdrak M, Kumrić M, Kurir TT, Božić J. Emerging Biomarkers for Early Detection of Chronic Kidney Disease. J Pers Med. 2022 Mar 31;12(4).
  30. Delgado C, Baweja M, Crews DC, Eneanya ND, Gadegbeku CA, Inker LA, et al. A Unifying Approach for GFR Estimation: Recommendations of the NKF-ASN Task Force on Reassessing the Inclusion of Race in Diagnosing Kidney Disease. Am J Kidney Dis. 2022 Feb;79(2):268-88 e1.
  31. Gottlieb ER, Estiverne C, Tolan NV, Melanson SEF, Mendu ML. Estimated GFR With Cystatin C and Creatinine in Clinical Practice: A Retrospective Cohort Study. Kidney Med. 2023 Mar;5(3):100600.
  32. Levey AS, Coresh J, Greene T, Stevens LA, Zhang YL, Hendriksen S, et al. Using standardized serum creatinine values in the modification of diet in renal disease study equation for estimating glomerular filtration rate. Ann Intern Med. 2006 Aug 15;145(4):247-54.
  33. Schwartz GJ, Muñoz A, Schneider MF, Mak RH, Kaskel F, Warady BA, et al. New equations to estimate GFR in children with CKD. J Am Soc Nephrol. 2009 Mar;20(3):629-37.
  34. Inker LA, Schmid CH, Tighiouart H, Eckfeldt JH, Feldman HI, Greene T, et al. Estimating glomerular filtration rate from serum creatinine and cystatin C. N Engl J Med. 2012 Jul 5;367(1):20-9.
  35. Pierce CB, Muñoz A, Ng DK, Warady BA, Furth SL, Schwartz GJ. Age- and sex-dependent clinical equations to estimate glomerular filtration rates in children and young adults with chronic kidney disease. Kidney Int. 2021 Apr;99(4):948-56.
  36. Inker LA, Tighiouart H, Adingwupu OM, Ng DK, Estrella MM, Maahs D, et al. Performance of GFR Estimating Equations in Young Adults. Am J Kidney Dis. 2024 Feb;83(2):272-76.
  37. Halacova M, Kotaska K, Kukacka J, Vavrova V, Kuzelova M, Ticha J, et al. Serum cystatin C level for better assessment of glomerular filtration rate in cystic fibrosis patients treated by amikacin. J Clin Pharm Ther. 2008 Aug;33(4):409-17.
  38. Beringer PM, Hidayat L, Heed A, Zheng L, Owens H, Benitez D, et al. GFR estimates using cystatin C are superior to serum creatinine in adult patients with cystic fibrosis. J Cyst Fibros. 2009 Jan;8(1):19-25.
  39. Wallace A, Price A, Fleischer E, Khoury M, Filler G. Estimation of GFR in Patients With Cystic Fibrosis: A Cross-Sectional Study. Can J Kidney Health Dis. 2020;7:2054358119899312.
  40. Sung KC, Ryu S, Lee JY, Lee SH, Cheong E, Hyun YY, et al. Urine Albumin/Creatinine Ratio Below 30 mg/g is a Predictor of Incident Hypertension and Cardiovascular Mortality. J Am Heart Assoc. 2016 Sep 13;5(9).
  41. Chaumont A, De Winter F, Dumont X, Haufroid V, Bernard A. The threshold level of urinary cadmium associated with increased urinary excretion of retinol-binding protein and beta 2-microglobulin: a re-assessment in a large cohort of nickel-cadmium battery workers. Occup Environ Med. 2011 Apr;68(4):257-64.
  42. Telser A, Farbman AI, Chacko C. A low-molecular-weight soluble protein from bovine lingual epithelium. II. Purification and characterization. J Invest Dermatol. 1982 Nov;79(5):286-92.
  43. Tate J, Caldwell G, Daly J, Gillis D, Jenkins M, Jovanovich S, et al. Recommendations for standardized reporting of protein electrophoresis in Australia and New Zealand. Ann Clin Biochem. 2012 May;49(Pt 3):242-56.
  44. Zhang J, Lou X, Shen H, Zellmer L, Sun Y, Liu S, et al. Isoforms of wild type proteins often appear as low molecular weight bands on SDS-PAGE. Biotechnol J. 2014 Aug;9(8):1044-54.
  45. Hamadah A, Gharaibeh K. Fractional Excretion of Sodium and Urea are Useful Tools in the Evaluation of AKI: PRO. Kidney360. 2023 Jun 1;4(6):e725-e27.
  46. Wołyniec W, Kasprowicz K, Rita-Tkachenko P, Renke M, Ratkowski W. Biochemical Markers of Renal Hypoperfusion, Hemoconcentration, and Proteinuria after Extreme Physical Exercise. Medicina (Kaunas). 2019 May 17;55(5).
  47. Levey AS, Coresh J. Chronic kidney disease. Lancet. 2012 Jan 14;379(9811):165-80.
  48. Dabla PK. Renal function in diabetic nephropathy. World J Diabetes. 2010 May 15;1(2):48-56.
  49. Perazella MA, Markowitz GS. Drug-induced acute interstitial nephritis. Nat Rev Nephrol. 2010 Aug;6(8):461-70.
  50. Real de Asúa D, Costa R, Galván JM, Filigheddu MT, Trujillo D, Cadiñanos J. Systemic AA amyloidosis: epidemiology, diagnosis, and management. Clin Epidemiol. 2014;6:369-77.
  51. Scurati-Manzoni E, Fossali EF, Agostoni C, Riva E, Simonetti GD, Zanolari-Calderari M, et al. Electrolyte abnormalities in cystic fibrosis: systematic review of the literature. Pediatr Nephrol. 2014 Jun;29(6):1015-23.
  52. Moresco RN, Speeckaert MM, Delanghe JR. Diagnosis and monitoring of IgA nephropathy: the role of biomarkers as an alternative to renal biopsy. Autoimmun Rev. 2015 Oct;14(10):847-53.
  53. Chapter 30 - Kidney Stones. In: Shlomo Melmed and Kenneth SPaPRLaHMK, editor. Williams Textbook of Endocrinology (Thirteenth Edition). Philadelphia: Elsevier; 2016. p. 1365-84.
  54. Downes KJ, Goldstein SL. Biomarkers for Kidney Injury in Cystic Fibrosis. Biomarkers in Kidney Disease. Dordrecht: Springer Netherlands; 2016. p. 689-718.
  55. Joyce E, Glasner P, Ranganathan S, Swiatecka-Urban A. Tubulointerstitial nephritis: diagnosis, treatment, and monitoring. Pediatr Nephrol. 2017 Apr;32(4):577-87.
  56. Webster AC, Nagler EV, Morton RL, Masson P. Chronic Kidney Disease. Lancet. 2017 Mar 25;389(10075):1238-52.
  57. Fan H, Hui L, Yan X, Hou W, Bai E, Wang L, et al. Serum 25 hydroxyvitamin D levels and affecting factors among preconception fertile women. BMC Womens Health. 2020 Jul 16;20(1):146.
  58. Rasheda Amin and Sun-Young Ahn and Asha M. Chapter 7 - Kidney and urinary tract disorders. In: Dennis Dietzen and Michael Bennett and Edward Wong and Shannon H, editor. Biochemical and Molecular Basis of Pediatric Disease (Fifth Edition). Academic Press; 2021. p. 167-228.
  59. Allinovi M, Trivioli G, Gaudio C, L'Imperio V, Rauf MU, Gillmore JD. The evolving spectrum of kidney amyloidosis: advances in diagnosis, typing and treatment. Nephrol Dial Transplant. 2025 Feb 26.
  60. Meher D, Agarwal V, Das S, Choudhury A, Sahoo D, Sahu SK, et al. Idiopathic Hypercalciuria: A Comprehensive Review of Clinical Insights and Management Strategies. Cureus. 2025 Apr;17(4):e81778.
  61. Alge JL, Arthur JM. Biomarkers of AKI: a review of mechanistic relevance and potential therapeutic implications. Clin J Am Soc Nephrol. 2015 Jan 7;10(1):147-55.
  62. Administration USFD. Letter of support for Drug-Induced Kidney Injurt (DIKI) Renal Tubular Injury Biomarkers. Letter. 2016.
  63. Han WK, Bailly V, Abichandani R, Thadhani R, Bonventre JV. Kidney Injury Molecule-1 (KIM-1): a novel biomarker for human renal proximal tubule injury. Kidney Int. 2002 Jul;62(1):237-44.
  64. Bonventre, JV. Kidney Injury Molecule-1 (KIM-1): a specific and sensitive biomarker of kidney injury. Scand J Clin Lab Invest Suppl. 2008;241:78-83.
  65. Bonventre, JV. Kidney injury molecule-1 (KIM-1): a urinary biomarker and much more. Nephrol Dial Transplant. 2009 Nov;24(11):3265-8.
  66. Lin X, Yuan J, Zhao Y, Zha Y. Urine interleukin-18 in prediction of acute kidney injury: a systemic review and meta-analysis. J Nephrol. 2015 Feb;28(1):7-16.
  67. Shang W, Wang Z. The Update of NGAL in Acute Kidney Injury. Curr Protein Pept Sci. 2017;18(12):1211-17.
  68. Rosner GM, Goswami HB, Sessions K, Mendyka LK, Kerin B, Vlasac I, et al. Lung-kidney axis in cystic fibrosis: Early urinary markers of kidney injury correlate with neutrophil activation and worse lung function. J Cyst Fibros. 2025 Jan 2.
  69. Huang F, Zeng Y, Lv L, Chen Y, Yan Y, Luo L, et al. Predictive value of urinary cell cycle arrest biomarkers for all cause-acute kidney injury: a meta-analysis. Sci Rep. 2023 Apr 13;13(1):6037.
  70. Menon V, Shlipak MG, Wang X, Coresh J, Greene T, Stevens L, et al. Cystatin C as a risk factor for outcomes in chronic kidney disease. Ann Intern Med. 2007 Jul 3;147(1):19-27.
  71. Bolignano D, Lacquaniti A, Coppolino G, Donato V, Campo S, Fazio MR, et al. Neutrophil gelatinase-associated lipocalin (NGAL) and progression of chronic kidney disease. Clin J Am Soc Nephrol. 2009 Feb;4(2):337-44.
  72. Duru OK, Vargas RB, Kermah D, Nissenson AR, Norris KC. High prevalence of stage 3 chronic kidney disease in older adults despite normal serum creatinine. J Gen Intern Med. 2009 Jan;24(1):86-92.
  73. Porazko T, Kúzniar J, Kusztal M, Kúzniar TJ, Weyde W, Kuriata-Kordek M, et al. IL-18 is involved in vascular injury in end-stage renal disease patients. Nephrol Dial Transplant. 2009 Feb;24(2):589-96.
  74. Waikar SS, Bonventre JV. Creatinine kinetics and the definition of acute kidney injury. J Am Soc Nephrol. 2009 Mar;20(3):672-9.
  75. Jose Luis Gorriz and Alberto M-C. Proteinuria: detection and role in native renal disease progression. Transplantation Reviews. 2012;26(1):3-13.
  76. Mula-Abed WA, Al Rasadi K, Al-Riyami D. Estimated Glomerular Filtration Rate (eGFR): A Serum Creatinine-Based Test for the Detection of Chronic Kidney Disease and its Impact on Clinical Practice. Oman Med J. 2012 Mar;27(2):108-13.
  77. Ramseyer VD, Garvin JL. Tumor necrosis factor-α: regulation of renal function and blood pressure. Am J Physiol Renal Physiol. 2013 May 15;304(10):F1231-42.
  78. Yong K, Ooi EM, Dogra G, Mannion M, Boudville N, Chan D, et al. Elevated interleukin-12 and interleukin-18 in chronic kidney disease are not associated with arterial stiffness. Cytokine. 2013 Oct;64(1):39-42.
  79. Lee BT, Ahmed FA, Hamm LL, Teran FJ, Chen CS, Liu Y, et al. Association of C-reactive protein, tumor necrosis factor-alpha, and interleukin-6 with chronic kidney disease. BMC Nephrol. 2015 May 30;16:77.
  80. Sedighi O, Abediankenari S, Omranifar B. Association between plasma Beta-2 microglobulin level and cardiac performance in patients with chronic kidney disease. Nephrourol Mon. 2015 Jan;7(1):e23563.
  81. Argyropoulos CP, Chen SS, Ng YH, Roumelioti ME, Shaffi K, Singh PP, et al. Rediscovering Beta-2 Microglobulin As a Biomarker across the Spectrum of Kidney Diseases. Front Med (Lausanne). 2017;4:73.
  82. Seki M, Nakayama M, Sakoh T, Yoshitomi R, Fukui A, Katafuchi E, et al. Blood urea nitrogen is independently associated with renal outcomes in Japanese patients with stage 3-5 chronic kidney disease: a prospective observational study. BMC Nephrol. 2019 Apr 2;20(1):115.
  83. Song J, Yu J, Prayogo GW, Cao W, Wu Y, Jia Z, et al. Understanding kidney injury molecule 1: a novel immune factor in kidney pathophysiology. Am J Transl Res. 2019;11(3):1219-29.
  84. Benoit SW, Ciccia EA, Devarajan P. Cystatin C as a biomarker of chronic kidney disease: latest developments. Expert Rev Mol Diagn. 2020 Oct;20(10):1019-26.
  85. Laville SM, Couturier A, Lambert O, Metzger M, Mansencal N, Jacquelinet C, et al. Urea levels and cardiovascular disease in patients with chronic kidney disease. Nephrol Dial Transplant. 2022 Feb 26;38(1):184-92.
  86. Azukaitis K, Ju W, Kirchner M, Nair V, Smith M, Fang Z, et al. Low levels of urinary epidermal growth factor predict chronic kidney disease progression in children. Kidney Int. 2019 Jul;96(1):214-21.
  87. Menez S, Wen Y, Xu L, Moledina DG, Thiessen-Philbrook H, Hu D, et al. The ASSESS-AKI Study found urinary epidermal growth factor is associated with reduced risk of major adverse kidney events. Kidney Int. 2023 Dec;104(6):1194-205.
  88. Guy EL, Bosomworth M, Denton M, Conway SP, Brownlee KG, Lee TW. Serum tobramycin levels following delivery of tobramycin (Tobi) via eFlow advanced nebuliser in children with cystic fibrosis. J Cyst Fibros. 2010 Jul;9(4):292-5.
  89. van Velzen AJ, Uges JWF, Heijerman HGM, Arets BGM, Nuijsink M, van der Wiel-Kooij EC, et al. Pharmacokinetics and safety of tobramycin nebulization with the I-neb and PARI-LC Plus in children with cystic fibrosis: A randomized, crossover study. Br J Clin Pharmacol. 2019 Sep;85(9):1984-93.
  90. Hart A, Cesar F, Zelnick LR, O'Connor N, Bailey Z, Lo J, et al. Identification of prognostic biomarkers for antibiotic associated nephrotoxicity in cystic fibrosis. J Cyst Fibros. 2024 Mar;23(2):293-99.
  91. Grange C, Bussolati B. Extracellular vesicles in kidney disease. Nature Reviews Nephrology. 2022 2022/08/01;18(8):499-513.
  92. Quesenberry PJ, Aliotta J, Deregibus MC, Camussi G. Role of extracellular RNA-carrying vesicles in cell differentiation and reprogramming. Stem Cell Res Ther. 2015 Sep 3;6:153.
  93. Grange C, Bussolati B. Extracellular vesicles in kidney disease. Nat Rev Nephrol. 2022 Aug;18(8):499-513.
  94. Yavuz H, Weder MM, Erdbrügger U. Extracellular Vesicles in Acute Kidney Injury. Nephron. 2023;147(1):48-51.
  95. Hessvik NP, Llorente A. Current knowledge on exosome biogenesis and release. Cell Mol Life Sci. 2018 Jan;75(2):193-208.
  96. Pisitkun T, Shen RF, Knepper MA. Identification and proteomic profiling of exosomes in human urine. Proc Natl Acad Sci U S A. 2004 Sep 7;101(36):13368-73.
  97. Wu Q, Poulsen SB, Murali SK, Grimm PR, Su XT, Delpire E, et al. Large-Scale Proteomic Assessment of Urinary Extracellular Vesicles Highlights Their Reliability in Reflecting Protein Changes in the Kidney. J Am Soc Nephrol. 2021 Sep;32(9):2195-209.
  98. Erdbrugger U, Hoorn EJ, Le TH, Blijdorp CJ, Burger D. Extracellular Vesicles in Kidney Diseases: Moving Forward. Kidney360. 2023 Feb 1;4(2):245-57.
  99. Alvarez S, Suazo C, Boltansky A, Ursu M, Carvajal D, Innocenti G, et al. Urinary exosomes as a source of kidney dysfunction biomarker in renal transplantation. Transplant Proc. 2013;45(10):3719-23.
  100. Panich T, Chancharoenthana W, Somparn P, Issara-Amphorn J, Hirankarn N, Leelahavanichkul A. Urinary exosomal activating transcriptional factor 3 as the early diagnostic biomarker for sepsis-induced acute kidney injury. BMC Nephrol. 2017 Jan 7;18(1):10.
  101. Miller D, Eagle-Hemming B, Sheikh S, Joel-David L, Adebayo A, Lai FY, et al. Urinary extracellular vesicles and micro-RNA as markers of acute kidney injury after cardiac surgery. Sci Rep. 2022 Jun 21;12(1):10402.
  102. Svenningsen P, Maslauskiene R, Palarasah Y, Bumblyte IA, Tepel M. Urinary Extracellular Vesicles for Non-Invasive Quantification of Principal Cell Damage in Kidney Transplant Recipients. Biomolecules. 2024 Sep 5;14(9).
  103. Dimuccio V, Peruzzi L, Brizzi MF, Cocchi E, Fop F, Boido A, et al. Acute and chronic glomerular damage is associated with reduced CD133 expression in urinary extracellular vesicles. Am J Physiol Renal Physiol. 2020 Feb 1;318(2):F486-f95.
  104. Ranghino A, Dimuccio V, Papadimitriou E, Bussolati B. Extracellular vesicles in the urine: markers and mediators of tissue damage and regeneration. Clin Kidney J. 2015 Feb;8(1):23-30.
  105. Santelli A, Sun IO, Eirin A, Abumoawad AM, Woollard JR, Lerman A, et al. Senescent Kidney Cells in Hypertensive Patients Release Urinary Extracellular Vesicles. J Am Heart Assoc. 2019 Jun 4;8(11):e012584.
  106. Hamasaki and Masataka Hisano and Tae Omori and Takayuki Okamoto and Hirotsugu Kitayama and Naoya Fujita and Hiromi Kuramochi and Takanori Ichiki and Akira Oka and Yutaka, Harita KTaKUaMSaENaTNaYKaSKaKMaMHaJHaY. Urinary extracellular vesicles signature for diagnosis of kidney disease. iScience. 2022;25(11):105416.
  107. Sepahzad A, Morris-Rosendahl DJ, Davies JC. Cystic Fibrosis Lung Disease Modifiers and Their Relevance in the New Era of Precision Medicine. Genes (Basel). 2021 Apr 13;12(4).
  108. Deltas C, Papagregoriou G, Louka SF, Malatras A, Flinter F, Gale DP, et al. Genetic Modifiers of Mendelian Monogenic Collagen IV Nephropathies in Humans and Mice. Genes (Basel). 2023 Aug 25;14(9).
  109. Leggatt GP, Seaby EG, Veighey K, Gast C, Gilbert RD, Ennis S. A Role for Genetic Modifiers in Tubulointerstitial Kidney Diseases. Genes (Basel). 2023 Aug 3;14(8).
  110. Zhou YH, Gallins PJ, Pace RG, Dang H, Aksit MA, Blue EE, et al. Genetic Modifiers of Cystic Fibrosis Lung Disease Severity: Whole-Genome Analysis of 7,840 Patients. Am J Respir Crit Care Med. 2023 May 15;207(10):1324-33.
  111. Esnault VL, Testa A, Audrain M, Roge C, Hamidou M, Barrier JH, et al. Alpha 1-antitrypsin genetic polymorphism in ANCA-positive systemic vasculitis. Kidney Int. 1993 Jun;43(6):1329-32.
  112. Doring G, Krogh-Johansen H, Weidinger S, Hoiby N. Allotypes of alpha 1-antitrypsin in patients with cystic fibrosis, homozygous and heterozygous for deltaF508. Pediatr Pulmonol. 1994 Jul;18(1):3-7.
  113. Hull J, Thomson AH. Contribution of genetic factors other than CFTR to disease severity in cystic fibrosis. Thorax. 1998 Dec;53(12):1018-21.
  114. Mahadeva R, Westerbeek RC, Perry DJ, Lovegrove JU, Whitehouse DB, Carroll NR, et al. Alpha1-antitrypsin deficiency alleles and the Taq-I G-->A allele in cystic fibrosis lung disease. Eur Respir J. 1998 Apr;11(4):873-9.
  115. Tanaka R, Iijima K, Murakami R, Koide M, Nakamura H, Yoshikawa N. ACE gene polymorphism in childhood IgA nephropathy: association with clinicopathologic findings. Am J Kidney Dis. 1998 May;31(5):774-9.
  116. Allen RD. Polymorphism of the human TNF-alpha promoter--random variation or functional diversity? Mol Immunol. 1999 Oct-Nov;36(15-16):1017-27.
  117. Garred P, Pressler T, Madsen HO, Frederiksen B, Svejgaard A, Hoiby N, et al. Association of mannose-binding lectin gene heterogeneity with severity of lung disease and survival in cystic fibrosis. J Clin Invest. 1999 Aug;104(4):431-7.
  118. Grasemann H, Knauer N, Buscher R, Hubner K, Drazen JM, Ratjen F. Airway nitric oxide levels in cystic fibrosis patients are related to a polymorphism in the neuronal nitric oxide synthase gene. Am J Respir Crit Care Med. 2000 Dec;162(6):2172-6.
  119. Henry MT, Cave S, Rendall J, O'Connor CM, Morgan K, FitzGerald MX, et al. An alpha1-antitrypsin enhancer polymorphism is a genetic modifier of pulmonary outcome in cystic fibrosis. Eur J Hum Genet. 2001 Apr;9(4):273-8.
  120. Bennett WD. Effect of beta-adrenergic agonists on mucociliary clearance. J Allergy Clin Immunol. 2002 Dec;110(6 Suppl):S291-7.
  121. Grasemann H, Storm van's Gravesande K, Gartig S, Kirsch M, Buscher R, Drazen JM, et al. Nasal nitric oxide levels in cystic fibrosis patients are associated with a neuronal NO synthase (NOS1) gene polymorphism. Nitric Oxide. 2002 Mar;6(2):236-41.
  122. Meyer P, Braun A, Roscher AA. Analysis of the two common alpha-1-antitrypsin deficiency alleles PiMS and PiMZ as modifiers of Pseudomonas aeruginosa susceptibility in cystic fibrosis. Clin Genet. 2002 Oct;62(4):325-7.
  123. Frangolias DD, Ruan J, Wilcox PJ, Davidson AG, Wong LT, Berthiaume Y, et al. Alpha 1-antitrypsin deficiency alleles in cystic fibrosis lung disease. Am J Respir Cell Mol Biol. 2003 Sep;29(3 Pt 1):390-6.
  124. Grasemann H, Storm van's Gravesande K, Buscher R, Knauer N, Silverman ES, Palmer LJ, et al. Endothelial nitric oxide synthase variants in cystic fibrosis lung disease. Am J Respir Crit Care Med. 2003 Feb 1;167(3):390-4.
  125. Bantis C, Heering PJ, Aker S, Klein-Vehne N, Grabensee B, Ivens K. Association of interleukin-10 gene G-1082A polymorphism with the progression of primary glomerulonephritis. Kidney Int. 2004 Jul;66(1):288-94.
  126. Flamant C, Henrion-Caude A, Boelle PY, Bremont F, Brouard J, Delaisi B, et al. Glutathione-S-transferase M1, M3, P1 and T1 polymorphisms and severity of lung disease in children with cystic fibrosis. Pharmacogenetics. 2004 May;14(5):295-301.
  127. Jaber BL, Liangos O, Pereira BJ, Balakrishnan VS. Polymorphism of immunomodulatory cytokine genes: implications in acute renal failure. Blood Purif. 2004;22(1):101-11.
  128. Texereau J, Marullo S, Hubert D, Coste J, Dusser DJ, Dall'Ava-Santucci J, et al. Nitric oxide synthase 1 as a potential modifier gene of decline in lung function in patients with cystic fibrosis. Thorax. 2004 Feb;59(2):156-8.
  129. Wooldridge JL, Deutsch GH, Sontag MK, Osberg I, Chase DR, Silkoff PE, et al. NO pathway in CF and non-CF children. Pediatr Pulmonol. 2004 Apr;37(4):338-50.
  130. Zheng S, Xu W, Bose S, Banerjee AK, Haque SJ, Erzurum SC. Impaired nitric oxide synthase-2 signaling pathway in cystic fibrosis airway epithelium. Am J Physiol Lung Cell Mol Physiol. 2004 Aug;287(2):L374-81.
  131. Brouard J, Knauer N, Boelle PY, Corvol H, Henrion-Caude A, Flamant C, et al. Influence of interleukin-10 on Aspergillus fumigatus infection in patients with cystic fibrosis. J Infect Dis. 2005 Jun 1;191(11):1988-91.
  132. Yarden J, Radojkovic D, De Boeck K, Macek M, Jr., Zemkova D, Vavrova V, et al. Association of tumour necrosis factor alpha variants with the CF pulmonary phenotype. Thorax. 2005 Apr;60(4):320-5.
  133. Steagall WK, Barrow BJ, Glasgow CG, Mendoza JW, Ehrmantraut M, Lin JP, et al. Beta-2-adrenergic receptor polymorphisms in cystic fibrosis. Pharmacogenet Genomics. 2007 Jun;17(6):425-30.
  134. Li X, Magenheimer BS, Xia S, Johnson T, Wallace DP, Calvet JP, et al. A tumor necrosis factor-alpha-mediated pathway promoting autosomal dominant polycystic kidney disease. Nat Med. 2008 Aug;14(8):863-8.
  135. Pirson Y. Does TNF-alpha enhance cystogenesis in ADPKD? Nephrol Dial Transplant. 2008 Dec;23(12):3773-5.
  136. Tesse R, Cardinale F, Santostasi T, Polizzi A, Mappa L, Manca A, et al. Association of interleukin-10 gene haplotypes with Pseudomonas aeruginosa airway colonization in cystic fibrosis. J Cyst Fibros. 2008 Jul;7(4):329-32.
  137. Brand P, Schulte M, Wencker M, Herpich CH, Klein G, Hanna K, et al. Lung deposition of inhaled alpha1-proteinase inhibitor in cystic fibrosis and alpha1-antitrypsin deficiency. Eur Respir J. 2009 Aug;34(2):354-60.
  138. Vuong MT, Lundberg S, Gunnarsson I, Wramner L, Seddighzadeh M, Hahn-Zoric M, et al. Genetic variation in the transforming growth factor-beta1 gene is associated with susceptibility to IgA nephropathy. Nephrol Dial Transplant. 2009 Oct;24(10):3061-7.
  139. Baylis C. Nitric oxide synthase derangements and hypertension in kidney disease. Curr Opin Nephrol Hypertens. 2012 Jan;21(1):1-6.
  140. Marson FA, Bertuzzo CS, Hortencio TD, Ribeiro JD, Bonadia LC, Ribeiro AF. The ACE gene D/I polymorphism as a modulator of severity of cystic fibrosis. BMC Pulm Med. 2012 Aug 8;12:41.
  141. Sinuani I, Beberashvili I, Averbukh Z, Sandbank J. Role of IL-10 in the progression of kidney disease. World J Transplant. 2013 Dec 24;3(4):91-8.
  142. Susantitaphong P, Perianayagam MC, Tighiouart H, Liangos O, Bonventre JV, Jaber BL. Tumor necrosis factor alpha promoter polymorphism and severity of acute kidney injury. Nephron Clin Pract. 2013;123(1-2):67-73.
  143. Sanchez-Dominguez CN, Reyes-Lopez MA, Bustamante A, Cerda-Flores RM, Villalobos-Torres Mdel C, Gallardo-Blanco HL, et al. The tumor necrosis factor alpha (-308 A/G) polymorphism is associated with cystic fibrosis in Mexican patients. PLoS One. 2014;9(3):e90945.
  144. Zager RA, Johnson AC, Frostad KB. Rapid renal alpha-1 antitrypsin gene induction in experimental and clinical acute kidney injury. PLoS One. 2014;9(5):e98380.
  145. McElvaney NG. Alpha-1 Antitrypsin Therapy in Cystic Fibrosis and the Lung Disease Associated with Alpha-1 Antitrypsin Deficiency. Ann Am Thorac Soc. 2016 Apr;13 Suppl 2:S191-6.
  146. Nomani H, Hagh-Nazari L, Aidy A, Vaisi-Raygani A, Kiani A, Rahimi Z, et al. Association between GSTM1, GSTT1, and GSTP1 variants and the risk of end stage renal disease. Ren Fail. 2016 Oct;38(9):1455-61.
  147. Sarkar T, Singh NP, Kar P, Husain SA, Kapoor S, Pollipalli SK, et al. Does angiotensin-converting enzyme-1 (ACE-1) gene polymorphism lead to chronic kidney disease among hypertensive patients? Ren Fail. 2016 Jun;38(5):765-9.
  148. Vijftigschild LA, Berkers G, Dekkers JF, Zomer-van Ommen DD, Matthes E, Kruisselbrink E, et al. beta2-Adrenergic receptor agonists activate CFTR in intestinal organoids and subjects with cystic fibrosis. Eur Respir J. 2016 Sep;48(3):768-79.
  149. Guo WY, Zhu L, Meng SJ, Shi SF, Liu LJ, Lv JC, et al. Mannose-Binding Lectin Levels Could Predict Prognosis in IgA Nephropathy. J Am Soc Nephrol. 2017 Nov;28(11):3175-81.
  150. Chan SC, Zhang Y, Shao A, Avdulov S, Herrera J, Aboudehen K, et al. Mechanism of Fibrosis in HNF1B-Related Autosomal Dominant Tubulointerstitial Kidney Disease. J Am Soc Nephrol. 2018 Oct;29(10):2493-509.
  151. Medina AM, Zubero EE, Jimenez MAA, Barragan SAA, Garcia CAL, Ramos JJG, et al. NOS3 Polymorphisms and Chronic Kidney Disease. J Bras Nefrol. 2018 Jul-Sep;40(3):273-77.
  152. Arif E, Nihalani D. Beta2-adrenergic receptor in kidney biology: A current prospective. Nephrology (Carlton). 2019 May;24(5):497-503.
  153. Du S, Tian J, Xiao Z, Luo Z, Lin T, Zheng S, et al. Serum alpha 1-antitrypsin predicts severe acute kidney injury after cardiac surgery. J Thorac Dis. 2019 Dec;11(12):5053-62.
  154. Fernandez-Ruiz M, Gimenez E, Lora D, Aguado JM, Pascual M, Manuel O. Impact of MBL2 gene polymorphisms on the risk of infection in solid organ transplant recipients: A systematic review and meta-analysis. Am J Transplant. 2019 Apr;19(4):1072-85.
  155. Cai K, Ma Y, Wang J, Nie W, Guo J, Zhang M, et al. Mannose-binding lectin activation is associated with the progression of diabetic nephropathy in type 2 diabetes mellitus patients. Ann Transl Med. 2020 Nov;8(21):1399.
  156. Zhang Y, Dai Y, Raman A, Daniel E, Metcalf J, Reif G, et al. Overexpression of TGF-beta1 induces renal fibrosis and accelerates the decline in kidney function in polycystic kidney disease. Am J Physiol Renal Physiol. 2020 Dec 1;319(6):F1135-F48.
  157. Kamiar A, Yousefi K, Dunkley JC, Webster KA, Shehadeh LA. beta(2)-Adrenergic receptor agonism as a therapeutic strategy for kidney disease. Am J Physiol Regul Integr Comp Physiol. 2021 May 1;320(5):R575-R87.
  158. Mu H, Zheng Q, Hao L. IL-10 -1082 A/G polymorphism is related with the risk and clinical characteristics of acute kidney injury: a case-control study. BMC Nephrol. 2021 Jun 5;22(1):212.
  159. Santos GF, Ellis P, Farrugia D, Turner AM. Nephrotic syndrome secondary to alpha-1 antitrypsin deficiency. BMJ Case Rep. 2021 Mar 5;14(3).
  160. Tang PC, Chan AS, Zhang CB, Garcia Cordoba CA, Zhang YY, To KF, et al. TGF-beta1 Signaling: Immune Dynamics of Chronic Kidney Diseases. Front Med (Lausanne). 2021;8:628519.
  161. Baldassarri M, Zguro K, Tomati V, Pastorino C, Fava F, Croci S, et al. Gain- and Loss-of-Function CFTR Alleles Are Associated with COVID-19 Clinical Outcomes. Cells. 2022 Dec 16;11(24).
  162. Levy RV, Reidy KJ, Le TH, David V, Winkler C, Xu Y, et al. Association of GSTM1 Deletion With Progression of CKD in Children: Findings From the Chronic Kidney Disease in Children (CKiD) Study. Am J Kidney Dis. 2022 Jul;80(1):79-86.
  163. Padhi UN, Mulkalwar M, Saikrishna L, Verma HK, Bhaskar L. NOS3 gene intron 4 a/b polymorphism is associated with ESRD in autosomal dominant polycystic kidney disease patients. J Bras Nefrol. 2022 Apr-Jun;44(2):224-31.
  164. Trojan T, Alejandre Alcazar MA, Fink G, Thomassen JC, Maessenhausen MV, Rietschel E, et al. The effect of TGF-beta(1) polymorphisms on pulmonary disease progression in patients with cystic fibrosis. BMC Pulm Med. 2022 May 7;22(1):183.
  165. Barratt J, Lafayette RA, Zhang H, Tesar V, Rovin BH, Tumlin JA, et al. IgA nephropathy: the lectin pathway and implications for targeted therapy. Kidney Int. 2023 Aug;104(2):254-64.
  166. Bezzerri V, Gentili V, Api M, Finotti A, Papi C, Tamanini A, et al. SARS-CoV-2 viral entry and replication is impaired in Cystic Fibrosis airways due to ACE2 downregulation. Nat Commun. 2023 Jan 10;14(1):132.
  167. Lutful Kabir F, Ambalavanan N, Liu G, Li P, Solomon GM, Lal CV, et al. MicroRNA-145 Antagonism Reverses TGF-beta Inhibition of F508del CFTR Correction in Airway Epithelia. Am J Respir Crit Care Med. 2018 Mar 1;197(5):632-43.
  168. Sun H, Harris WT, Kortyka S, Kotha K, Ostmann AJ, Rezayat A, et al. Tgf-beta downregulation of distinct chloride channels in cystic fibrosis-affected epithelia. PloS one. 2014;9(9):e106842.
  169. Zhang Y, He D, Zhang W, Xing Y, Guo Y, Wang F, et al. ACE Inhibitor Benefit to Kidney and Cardiovascular Outcomes for Patients with Non-Dialysis Chronic Kidney Disease Stages 3-5: A Network Meta-Analysis of Randomised Clinical Trials. Drugs. 2020 Jun;80(8):797-811.
  170. Baylis, C. Nitric oxide deficiency in chronic kidney disease. Am J Physiol Renal Physiol. 2008 Jan;294(1):F1-9.
  171. Phua YL, Chen KH, Hemker SL, Marrone AK, Bodnar AJ, Liu X, et al. Loss of miR-17~92 results in dysregulation of Cftr in nephron progenitors. Am J Physiol Renal Physiol. 2019 May 1;316(5):F993-F1005.
  172. van Heeckeren AM, Schluchter MD, Drumm ML, Davis PB. Role of Cftr genotype in the response to chronic Pseudomonas aeruginosa lung infection in mice. American journal of physiology Lung cellular and molecular physiology. 2004 Nov;287(5):L944-52.
  173. Hodges CA, Grady BR, Mishra K, Cotton CU, Drumm ML. Cystic fibrosis growth retardation is not correlated with loss of Cftr in the intestinal epithelium. American journal of physiology Gastrointestinal and liver physiology. 2011 Sep;301(3):G528-36.
  174. McHugh DR, Steele MS, Valerio DM, Miron A, Mann RJ, LePage DF, et al. A G542X cystic fibrosis mouse model for examining nonsense mutation directed therapies. PloS one. 2018;13(6):e0199573.
  175. Sun X, Yi Y, Yan Z, Rosen BH, Liang B, Winter MC, et al. In utero and postnatal VX-770 administration rescues multiorgan disease in a ferret model of cystic fibrosis. Sci Transl Med. 2019 Mar 27;11(485).
  176. Birket SE, Davis JM, Fernandez-Petty CM, Henderson AG, Oden AM, Tang L, et al. Ivacaftor Reverses Airway Mucus Abnormalities in a Rat Model Harboring a Humanized G551D-CFTR. Am J Respir Crit Care Med. 2020 Nov 1;202(9):1271-82.
  177. Birket SE, Davis JM, Fernandez CM, Tuggle KL, Oden AM, Chu KK, et al. Development of an airway mucus defect in the cystic fibrosis rat. JCI Insight. 2018 Jan 11;3(1).
  178. Sun X, Sui H, Fisher JT, Yan Z, Liu X, Cho HJ, et al. Disease phenotype of a ferret CFTR-knockout model of cystic fibrosis. J Clin Invest. 2010 Sep;120(9):3149-60.
  179. Delrue C, De Bruyne S, Speeckaert MM. Application of Machine Learning in Chronic Kidney Disease: Current Status and Future Prospects. Biomedicines. 2024;12(3):568.
Table 1. eGFR formulas based on age groups and sex.
Table 1. eGFR formulas based on age groups and sex.
eGFR Formula Population Age Group Characteristics
CKiD U25 eGFR CKiD 1-25 years Age, sex, height, serum creatinine, and serum Cystatin C [35,36]
CKD-EPI 2021 (Creatinine) General adult population ≥ 18 years Age, sex, and serum creatinine [36]
MDRD Study CKD ≥ 18 years Age, sex, race, and serum creatinine [32]
Schwartz Formula Pediatric population (general) 1-18 years Height and serum creatinine [33]
CKD-EPI 2012 (Creatinine-Cystatin C) General adult population ≥ 18 years Age, sex, race, serum creatinine, and serum Cystatin C [34]
CKiD, chronic kidney disease in children; eGFR, estimated glomerular filtration rate; CKD-EPI, chronic kidney disease epidemiology collaboration; MDRD, modification of diet in renal disease; CKD, chronic kidney disease.
Table 2. Standard tests used in clinical practice for the diagnosis of common kidney complications in PwCF (modified from Hart et al.) [7].
Table 2. Standard tests used in clinical practice for the diagnosis of common kidney complications in PwCF (modified from Hart et al.) [7].
Kidney complication Laboratory/Imaging test for detection
AKI Serum creatinine, BUN, urinalysis, urine microscopy, FeNa, FeUrea, and urine output monitoring [54]
Pseudo-Bartter syndrome Serum electrolytes, venous blood gas, and urine electrolytes [51]
AA amyloidosis Serum amyloid A, kidney biopsy, and SAP scintigraphy [50,59]
IgA nephropathy Urinalysis, urine microscopy, UPCR, and kidney biopsy [52]
Diabetic glomerulopathy Urinalysis, UACR, eGFR, and kidney biopsy [48]
Tubulointerstitial nephritis Urinalysis, kidney ultrasound, and kidney biopsy [49,55]
CKD eGFR, UACR, UPCR, and urinalysis [56]
LMW proteinuria (non-glomerular) Urine β2MG, and UPEP [58]
Hypercalciuria/
nephrolithiasis/
nephrocalcinosis
UCaCR, 24-hour urinary analysis calcium excretion, urine microscopy, serum 25-hydroxyvitamin D, and kidney ultrasound [53,57,60]
AKI, acute kidney injury; BUN, blood urea nitrogen, FeNa, fractional excretion of sodium; FeUrea, fractional excretion of urea, AA amyloidosis, secondary amyloidosis; SAP, serum amyloid P component); IgA, immunoglobulin A; UPCR, urine protein to creatinine ratio; UACR, urine albumin to creatinine ratio; eGFR, estimated glomerular filtration rate; CKD, chronic kidney disease; LMW, low molecular weight; β2MG, beta-2 microglobulin; UPEP, urine protein electrophoresis; UCaCR, urine calcium to creatinine ratio.
Table 3. Kidney injury and inflammatory markers and tests across CKD stages. Levels of early kidney injury markers such as KIM-1, NGAL, and IL-18 increase first, while indicators such as serum creatinine, BUN, and Cystatin C increase in later stages.
Table 3. Kidney injury and inflammatory markers and tests across CKD stages. Levels of early kidney injury markers such as KIM-1, NGAL, and IL-18 increase first, while indicators such as serum creatinine, BUN, and Cystatin C increase in later stages.
Biomarker/Test Type Early Stage (Subclinical Injury) Intermediate Stage (Functional Decline) Late Stage (Established CKD)
Serum creatinine [72,74] Functional decline Normal Starts increasing High in late-stage CKD
BUN [82,85] Functional decline Normal Increases moderately High in severe CKD
Cystatin C [70,84] Functional decline Normal or slightly elevated Moderately increased High in severe CKD
eGFR [76] Functional decline Normal or even high Progressive decline Severely reduced in CKD
Proteinuria [75] Glomerular dysfunction Slight increase Increases significantly Severe elevation in CKD
KIM-1 [83] Tubular injury marker Early rise Decreases with chronicity Low but persists in CKD
β2MG [80,81] Glomerular dysfunction Normal or slightly elevated Moderately increased High in severe CKD
NGAL [71] Tubular injury marker Early and rapid increase Fluctuates with injury Persistent in progressive CKD
IL-18 [73,78] Inflammatory marker Early marker of tubular damage Moderate increase Can persist in CKD
TNFα [77,79] Inflammatory marker Low or normal Elevated due to chronic inflammation Persistently high in late CKD
CKD, chronic kidney disease; BUN, blood urea nitrogen; eGFR, estimated glomerular filtration rate; KIM-1, kidney injury molecule-1; β2MG, beta-2 microglobulin; NGAL, neutrophil gelatinase-associated lipocalin; IL-18, interleukin-18; TNFα, tumor necrosis factor α.
Table 4. Common genetic determinants in CF and CKD.
Table 4. Common genetic determinants in CF and CKD.
Modifier Role in PwCF Role in CKD
TGFβ1 The most established genetic modifier in CF. Several TGFβ1 polymorphisms[111–166 are associated with CF progression and P. aeruginosa infection [164]. Drives HNF1β-induced ADTKD [150]. Susceptibility to IgAN [138], polycystic kidney diseases [156], and CKD [160].
ACE The D/I polymorphism is associated with disease severity [140]. Expression and localization are controlled by CFTR [161,166]. DD genotype is a risk factor for CKD [147]. ID/DD genotypes are associated with chronic lesions, such as capsular adhesions or glomerulosclerosis and proteinuria in severe In severe IgAN [115].
MBL2 Decreased survival and increased susceptibility to infections to P. aeruginosa and worse lung functions [117,154]. The pathogenic variants are Gly54Asp (the B allele, rs1800450), Gly57Glu (the C allele, rs1800451), and Arg52Cys (the D allele, rs5030737), which together referred to as 0 allele [143]. Glomerular deposition of MBL has been consistently observed in kidney biopsy specimens in people with IgAN [149,165]. High serum levels are also associated with the development and progression of diabetic nephropathy [155].
AAT The most abundant proteinase inhibitor in the lung with anti-inflammatory effects. Genetic modifier protecting against disease progression [119,123,137,145]. M (normal), S (264Glu→Val) and Z (342Glu→Lys) [114]. Contradictory results for the effect of S and Z alleles [112,114,119,122]. Rapid rise of serum levels predicts AKI in experimental and clinical settings [144,153]. S and Z alleles were associated with high levels of the antigen of ANCA in Granulomatosis with polyangiitis [111]. In CKD, AAT has a protective effect [159].
β2AR Stimulation results in improved lung function [120]. The Gly16Glu27 genetic variant upregulates CFTR activity [133,148]. Expressed in proximal tubules, glomeruli, and podocytes [152]. Anti-inflammatory [157].
TNFα (-308 GA, rs1800629) polymorphism is associated with CF [143]. +691g ins/del polymorphic locus is associated with the severity of lung disease and. aeruginosa infection [132]. TNFα -308GA promoter polymorphism (rs1800629) that were associated with high TNFα transcription, CF and AKI severity [116,127,142]. High levels disrupt the localization of PC2 to the plasma membrane and primary cilia in ADPKD [134,135].
IL-10 Anti-inflammatory cytokine present at low levels in PwCF. The haplotype GCC/ACC is significantly associated with P. aeruginosa infection and CF severity [136]. A significant association was found between the −1082GG genotype and colonization with A. fumigatus and allergic bronchopulmonary aspergillosis [131]. Important role in normal physiology, AKI and CKD progression [141]. Polymorphisms are associated with AKI [127]. L-10 -1082 A/G polymorphism was associated with an increased risk of AKI [158] and primary glomerulonephritis [125].
NOS Low levels of exhaled NO [129,130]. NOS1 and NOS2 polymorphisms are associated with disease severity and inflammation [118,121]. G847T polymorphism in the NOS3 gene, is associated with high NO production had a slower decline in lung function [124,128]. Levels are reduced in CKD. NOS inhibition causes systemic and glomerular hypertension, glomerular ischemia, glomerulosclerosis, tubulointerstitial injury, and proteinuria [139]. Presence of the two NOS3 gene polymorphisms, Glu298Asp polymorphisms 4 b/a and -786T>C is a risk of ESKD in patients with CKD and ADPKD [151,163].
GST M1 (GSTM1) allele associated with worse lung disease [113]. GSTM3*B allele contributes to clinical severity in CF [126]. GSTM1, GSTT1, and GSTP1 polymorphisms are risk of ESKD [146]. GSTM1 deletion associated with more rapid progression of pediatric CKD [162].
TGFβ1, transforming growth factor β1; CF, cystic fibrosis; HNF1β, hepatocyte nuclear factor 1-beta; ADTKD, autosomal dominant tubulointerstitial kidney disease; IgAN, immunoglobulin A nephropathy; ACE, angiotensin-converting enzyme; CFTR, CF transmembrane conductance regulator; MBL2, mannose-binding lectin; AAT, α1-antitrypsin; AKI, acute kidney injury; ANCA, antineutrophil cytoplasm antibodies; CKD, chronic kidney disease; β2AR, beta-2-adrenergic receptor; TNFα, tumor necrosis factor α; PC2, polycystin 2; ADPKD, autosomal dominant polycystic kidney disease; IL-10, interleukin-10; PwCF, people with cystic fibrosis; NOS, nitric oxide synthase; NO, nitric oxide; ESKD, end-stage kidney disease; GST, glutathione S-transferase.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2026 MDPI (Basel, Switzerland) unless otherwise stated