Minireviews Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Nephrol. Mar 25, 2025; 14(1): 102381
Published online Mar 25, 2025. doi: 10.5527/wjn.v14.i1.102381
Transition from acute kidney injury to chronic kidney disease in liver cirrhosis patients: Current perspective
Sudheer Marrapu, Ramesh Kumar, Department of Gastroenterology, All India Institute of Medical Sciences, Patna 801507, India
ORCID number: Sudheer Marrapu (0009-0002-8209-8648); Ramesh Kumar (0000-0001-5136-4865).
Author contributions: Marrapu S analyzed the data and wrote the manuscript; Kumar R designed the study and wrote the manuscript; all authors have read and approved the final manuscript.
Conflict-of-interest statement: The authors have no conflict-of-interest to declare.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Ramesh Kumar, MD, Department of Gastroenterology, All India Institute of Medical Sciences, Phulwari Sharif, Patna 801507, India. docrameshkr@gmail.com
Received: October 16, 2024
Revised: December 22, 2024
Accepted: January 11, 2025
Published online: March 25, 2025
Processing time: 96 Days and 16.2 Hours

Abstract

In liver cirrhosis patients, acute kidney injury (AKI) is a common and severe complication associated with significant morbidity and mortality, often leading to chronic kidney disease (CKD). This progression reflects a complex interplay of renal and hepatic pathophysiology, with AKI acting as an initiator through maladaptive repair mechanisms. These mechanisms—such as tubular cell cycle arrest, inflammatory cascades, and fibrotic processes—are exacerbated by the hemodynamic and neurohormonal disturbances characteristic of cirrhosis. Following AKI episodes, persistent kidney dysfunction or acute kidney disease (AKD) often serves as a bridge to CKD. AKD represents a critical phase in renal deterioration, characterized by prolonged kidney injury that does not fully meet CKD criteria but exceeds the temporal scope of AKI. The progression from AKD to CKD is further influenced by recurrent AKI episodes, impaired renal autoregulation, and systemic comorbidities such as diabetes and metabolic dysfunction-associated steatotic liver disease, which compound kidney damage. The clinical management of AKI and CKD in cirrhotic patients requires a multidimensional approach that includes early identification of kidney injury, the application of novel biomarkers, and precision interventions. Recent evidence underscores the inadequacy of traditional biomarkers in predicting the AKI-to-CKD progression, necessitating novel biomarkers for early detection and intervention.

Key Words: Renal dysfunction; Acute kidney injury; Chronic kidney disease; Cirrhosis; Hepatorenal syndrome

Core Tip: In liver cirrhosis patients, acute kidney injury (AKI) frequently progresses to chronic kidney disease (CKD) through an intermediary stage known as acute kidney disease. This progression is driven by maladaptive repair processes, such as tubular cell cycle arrest and fibrosis, and exacerbated by cirrhosis-related hemodynamic and neurohormonal changes. Early detection of this AKI-to-CKD transition is crucial, and novel biomarkers and innovative imaging techniques, such as renal elastography, may present promising avenues for risk assessment and diagnosis. Integrating these tools into clinical practice, alongside targeted antifibrotic therapies and oxidative stress modulation, presents a promising strategy for managing renal complications in cirrhosis patients.



INTRODUCTION

Acute kidney injury (AKI) frequently occurs during the natural course of liver cirrhosis. The negative impact of AKI on cirrhosis patients is well-established and includes longer hospital stays, increased morbidity, and significantly higher rates of both in-hospital, short-term, and long-term mortality[1]. The prevalence of acute renal dysfunction in patients with cirrhosis ranges from 14% to 50%[1]. As the severity of liver disease increases, the incidence and risk of developing repeated episodes of AKI also rise. Moreover, patients who are discharged after an AKI episode have a higher risk of experiencing multiple AKI episodes and developing chronic kidney disease (CKD)[2,3]. Each AKI episode results in a reduction of renal reserve due to the loss of functional nephrons. Further, during the recovery phase, maladaptive repair mechanisms may trigger an inflammatory cascade, leading to interstitial fibrosis and eventually progressing to CKD[2]. The number of cirrhosis patients with CKD had increased from about 1% in 2005 to as high as 46.8% in 2019, reflecting the increasing incidence and prevalence of chronic liver disease (CLD) worldwide[4]. This paradigm shift signifies the convergence of important epidemiological factors common to both CLD and CKD, such as obesity, diabetes mellitus, hypertension, and metabolic dysfunction-associated steatotic liver disease (MASLD), besides the heightened risk of AKI-to-CKD progression in cirrhosis patients[5,6]. The transition from AKI to CKD in cirrhosis patients poses a significant clinical challenge, highlighting the urgent need for early detection, prompt intervention, and comprehensive management strategies. Figure 1 depicts factors associated with development of CKD in cirrhosis patients, with AKI playing and important role as initiator. CKD in cirrhosis patients has significant clinical implications, including an increased risk of recurrent AKI, refractory ascites, higher mortality, increased hospitalization rates, bacterial infections, higher liver transplantation (LT) rates, and reduced post-transplant survival[3,6]. However, data on CKD in CLD patients remains scarce. Hence, the present article will briefly discuss the AKI-to-CKD transition, the factors responsible for it, novel biomarkers for early identification, clinical impacts, and future therapies aimed at halting this progression from AKI to CKD.

Figure 1
Figure 1 Diagrammatic representation of factors associated with chronic kidney disease in liver cirrhosis patients. Acute kidney disease due to various causes can progressed to chronic kidney disease (CKD) in a significant proportion of cirrhosis patients. In addition, diabetes mellitus, hypertension and fatty liver disease, and certain etiological factors also increase the risk of CKD. AKI: Acute kidney injury; AKD: Acute kidney disease; CKD: Chronic kidney disease; DM: Diabetes mellitus; GI: Gastrointestinal; HBV: Hepatis B virus; HCV: Hepatitis C virus; HTN: Hypertension; HRS: Hepatorenal syndrome; MASLD: Metabolic dysfunction-associated steatotic liver disease.
AKI: THE INITIATOR

Among patients with cirrhosis and ascites, the overall prevalence of AKI is estimated to be around 14% to 50%, with many developing renal impairment upon hospital admission[1]. In a meta-analysis of 30 studies involving 18474 patients with cirrhosis, 5648 developed AKI, resulting in a pooled AKI incidence of 0.29 (95%CI: 0.28-0.30)[7]. In a meta-analysis focused on the incidence of AKI in acute-on chronic liver failure (ACLF) patients, Jiang et al[8] found it to be 0.41 (95%CI: 0.32-0.50). Another meta-analysis by Bai et al[9] found the incidence of AKI in cirrhosis patients admitted with acute gastrointestinal bleeding to be 0.21 (95%CI: 0.16-0.25). This broad variation in numbers occurs possibly because of heterogeneity in study populations and the use of diverse criteria to define renal dysfunction. Due to challenges in assessing renal function with accurate and less reliable creatinine-based assays, the International Club of Ascites revised its diagnostic approach in 2015, adopting the Kidney Disease: Improving Global Outcomes (KDIGO) 2012 criteria to define AKI in the context of cirrhosis, although some limitations persist[10]. Another recent meta-analysis by Lekakis et al[11], the largest to date on AKI incidence in cirrhotic patients, analyzed 5202232 participants from 73 studies, reporting an overall AKI incidence of 0.33 (95%CI: 0.29–0.38) and confirming that AKI is a common complication. Intensive care unit (ICU) patients had the highest AKI incidence at 0.61, while ACLF patients also showed significantly higher rates than general inpatients (0.43 vs 0.29). Studies using international classification of diseases-10 code reported lower AKI rates (0.16), while those employing KDIGO criteria showed higher rates (0.71)[11].

As cirrhosis progresses, renal and circulatory abnormalities not only predispose the kidneys to injury but also actively induce damage, a phenomenon known as hepatorenal physiology, potentially leading to conditions such as prerenal azotemia and acute tubular necrosis (ATN). Prerenal azotemia, driven by reduced blood flow, is reversible, while ATN involves structural damage to the kidney tubules. The combination of hemodynamic changes, nephrotoxins, and inflammation drives a continuum of renal dysfunction, where AKI may begin as a functional disorder but can progress to irreversible structural injury with sustained hypoperfusion. This underscores the importance of early diagnosis and intervention to prevent permanent renal damage[12]. A meta-analysis of 13 cohort studies found that patients with AKI have a substantially increased risk of developing CKD and end-stage renal disease (ESRD), with incidence rates of 25.8 and 8.6 per 100 person-years, respectively. Compared to those without AKI, these patients faced higher risks of CKD [hazard ratio (HR) = 8.8], ESRD (HR = 3.1), and mortality (HR = 2.0), indicating that AKI significantly impacts both renal and overall long-term outcomes[13]. This risk appears to be much higher in cirrhosis patients due to underlying hemodynamic impairments and neurohormonal changes, which lead to increased renal vasoconstriction, reduced renal perfusion, and decreased glomerular filtration, predisposing these individuals to recurrent AKI episodes and subsequent CKD. Cirrhosis patients discharged following an episode of AKI are at an increased risk of recurrent episodes of AKI, progression to CKD, dialysis dependency and mortality[3]. Bassegoda et al[6] determined the frequency of CKD in 409 patients with cirrhosis (168 with AKI and 241 without AKI). At three months, 25% of AKI survivors (24 of 97) had CKD, compared to only 1% (2 of 188) of patients without AKI (P < 0.0001). Both the severity and recovery patterns of AKI affect long-term kidney health and the progression from AKI to CKD. A study on AKI recovery patterns in 22000 patients revealed that most of them (91%) had stage 1 AKI, with 71% recovering within 2 days (fast recovery). By one year, 18.2% of all patients developed CKD, with AKI patients showing a higher incidence (31.8%) than non-AKI patients (15.5%, P < 0.001). For stage 1 AKI, the adjusted relative risk of progressing to CKD stage 3 or higher was 1.43 for fast recovery, 2.00 for intermediate recovery (3–10 days), and 2.65 for slow (> 10 days) or unknown recovery[14]. In a recent study on a nationwide United States cohort of hospitalised cirrhosis patients AKI stage 1B patients had a significantly higher risk of AKI progression [OR = 1.42 (95%CI: 1.14-1.72)] and 90-day mortality (27.2% vs 19.7%, P < 0.001) than AKI stage 1A patients[15]. A longer time to AKI-recovery appears to increase the risk of adverse renal outcomes. Patidar et al[16] found that the cumulative incidence of subsequent major adverse kidney events, including development of de-novo CKD stage ≥ 3, was 15%, 20%, and 29% for 0-2 days, 3-7 days, and > 7 days recovery groups in a large cohort of patients with cirrhosis and AKI (n = 5937), with 75% achieving AKI-recovery. However, AKI no-recovery occurs in more than half (57%) of critically ill cirrhosis patients. These patients are more likely to have grade 3 ACLF and significantly higher mortality risk (sub HR = 3.55)[17].

The progression from AKI to CKD through an intermediary stage called acute kidney disease (AKD) represents a continuum of renal dysfunction (Figure 2). AKI often initiates this cascade with an acute renal insult that, if unresolved, leads to persistent kidney injury. Persistent AKI, defined by kidney dysfunction lasting beyond 48 hours, is an important precursor to AKD and is associated with higher rates of progression to CKD[18,19]. Conversely, those with CKD experience higher rates of AKI. This dynamic nature of renal function is evident in how AKI, AKD, and CKD overlap, leading to the consideration of AKI and CKD as interconnected syndromes[20,21]. Both AKI and CKD share common risk factors and independent risk factors for increased morbidity, mortality, negative clinical impacts, and reduced life expectancy in cirrhosis patients. In summary, patients with cirrhosis are at risk of developing AKI, which is associated with increased risk of morbidity, mortality and a risk of progressing to CKD. Early detection of AKI is essential since prompt therapy can reverse the condition and improve the outcome.

Figure 2
Figure 2 Transition of acute kidney injury to chronic kidney disease in cirrhosis patients: Natural history and management strategies. The progression from acute kidney injury to chronic kidney disease represents a continuum of renal dysfunction that occurs through an intermediary stage called acute kidney disease. AKD: Acute kidney disease; AKI: Acute kidney injury; CKD: Chronic kidney disease; NAKI: Non-acute kidney injury; SLKT: Simultaneous liver and kidney transplantation.
AKD: THE PROGRESSOR

The concept of AKD is introduced as a key intermediary phase in the progression from AKI to CKD. Unlike AKI, which is defined by a rapid decline in kidney function over a brief period (typically 7 days), AKD encompasses a broader range of kidney injuries lasting up to three months, bridging the gap between AKI and CKD. This concept is essential because many patients do not fully recover from AKI, where the renal pathophysiologic processes remain functional, but also do not meet the criteria for CKD or AKI, necessitating a more comprehensive classification[22]. According to KDIGO 2012 guidelines, AKD is broadly classified to include patients with an estimated glomerular filtration rate (eGFR) below 60 mL/minute per 1.73 m², a glomerular filtration rate (GFR) decrease of 35% or more, or a serum creatinine increase greater than 50%, all lasting for less than three months[23]. However, for cirrhosis patients, the criteria differ slightly. The International Club of Ascites expert committee permits the use of baseline creatinine levels from up to three months earlier when diagnosing AKI. For AKD in cirrhosis, the definition includes either an eGFR below 60 mL/minute per 1.73 m² for less than three months or a serum creatinine increase of less than 50% in the same duration[24]. This tailored approach accounts for the unique renal diagnostic challenges presented by cirrhosis. AKD can develop either as an extension of AKI (AKI-AKD) or independently, even without an initial rapid onset of kidney impairment (non-acute kidney injury-AKD). In simple terms, AKD reflects scenarios where kidney function deteriorates over a period too prolonged for AKI but too short to be classified as CKD.

In a prospective study involving 272 cirrhosis patients, 80 patients (29.4%) developed AKD during follow-up. Of these, 42 patients (52.5%) recovered from their first episode of AKD, 16 (20%) experienced a second episode of AKD, and 36 (45.0%) died while experiencing AKD. Additionally, AKD progressed to CKD in 11 patients (13.8%), as compared to 2.1% in patients who did not develop AKD (P < 0.001). The 5-year survival rate was significantly lower in patients who developed AKD than in those who did not (34.8% vs 88.8%, P < 0.001). The significant negative impact of AKD on transplant-free survival was demonstrated regardless of the severity of cirrhosis at the time of inclusion. Besides age, GFR and mean arterial pressure (MAP) were identified as independent predictors of AKD in patients with cirrhosis. Therefore, these factors should be closely monitored in this patient population[25]. This study highlights the potentially reversible nature of AKD, suggesting that the continuum from AKI to CKD is neither obvious nor linear if appropriate intervention is implemented.

The clinical significance of AKD has been demonstrated in non-cirrhotic patients too[26,27]. James et al[26] indicated that the presence of AKD without AKI increased the risk of mortality and progression to CKD. A five-year observational study found that 47% of 232 patients who developed AKI during their ICU admission progressed to AKD by day 7. The cumulative incidence of CKD at the five-year follow-up was 30%. Multivariate analysis revealed that the risk of CKD in AKD patients was significantly higher in the first six months post-AKI, decreasing thereafter[28]. These findings emphasize that the trajectory of kidney recovery is crucial, with the lack of recovery being more significant than the presence of AKD itself in predicting the transition to CKD, further signifying the reversible nature of AKD. AKD often goes undetected if not properly recognized, despite its potential long-term impacts. Current management primarily focuses on the initial stages of AKI rather than the post-AKI period. Many patients may still exhibit subtle kidney dysfunction or structural abnormalities after recovery from AKI, qualifying for AKD[29]. Recognizing AKD is critical as it expands our understanding of kidney disease beyond AKI and CKD alone. It enables early identification of patients at elevated risk for CKD, providing a vital window for proactive measures to prevent further progression, as well as integrating structural damage markers into assessment criteria. However, recognition of AKD in clinical practice faces barriers such as unclear definitions, limited awareness, and lack of readily available and reliable biomarkers. Addressing these challenges requires standardized diagnostic criteria, enhanced clinician education, integration of novel biomarkers, and improved electronic health systems for trend analysis. In summary, AKD represents a pivotal transitional phase between AKI and CKD, offering a crucial window for proactive interventions to halt disease progression. Effective management of AKD demands collaborative efforts and targeted research to develop and implement strategies for timely identification and treatment, ultimately improving patient outcomes.

CKD: THE OUTCOME

In cirrhosis patients, CKD is defined as an eGFR of less than 60 mL/minute per 1.73 m² for over three months. Estimating eGFR remains challenging in cirrhosis patients; however, the 6-variable Modification of Diet in Renal Disease (MDRD6) formula, which includes factors such as serum creatinine, age, gender, albumin, blood urea nitrogen, and race, is acceptable. Notably, the MDRD6 can overestimate GFR, especially in patients with GFR below 40 mL/minute, highlighting the need for future studies to refine this estimation method. The diagnosis of CKD in cirrhosis patients does not require imaging as structural damage often precedes imaging changes. Renal biopsy poses risks of bleeding due to coagulopathy and technical difficulties due to large-volume ascites[30]. KDIGO classifies CKD into structural and functional types based on the presence of kidney injury. The former type 2 hepatorenal syndrome (HRS), now termed HRS-CKD, is classified as functional CKD, which could be reversible[31]. However, ongoing renal vasoconstriction in functional CKD can lead to structural changes, evolving into structural CKD. Patients with cirrhosis often face additional risks of structural CKD due to conditions such as diabetes mellitus, hypertension, MASLD, and atherosclerosis[32].

Prevalence of CKD in cirrhosis patients

In a prospective study of 818 cirrhosis patients, 61% and 32.8% developed AKI and CKD, respectively, during follow-up. The median time to the development of CKD was 173 days, and interestingly, 81.7% of those who developed CKD had a prior history of AKI[33]. In a five-year follow-up observational study by Orieux et al[28], the cumulative incidence of CKD at five years was 30%, primarily due to the trajectory of non-recovery from AKI in 70% of patients. Another study evaluating patients with cirrhosis found that 25% of those who survived an episode of AKI developed CKD at three months post-AKI, compared to only 1% of patients without AKI, establishing the key pathogenic role of AKI in the development of CKD[6]. In a large cross-sectional study of 7440 adult cirrhosis patients from Taiwan, CKD was present in approximately 46% of patients[34]. In a prospectively managed North American Consortium for the Study of End-Stage Liver Disease data, the prevalence of CKD was 46.8% in non-electively admitted patients, of which 68% had superimposed AKI[4]. A large retrospective analysis of 78640 patients awaiting LT by Cullaro et al[35] reported the prevalence of CKD at 7.8% in 2002 compared to 14.6% in 2017, suggesting a rising trend over time.

Mechanism of AKI to CKD transition

After AKI, the kidneys initiate repair mechanisms involving renal progenitor cells and de-differentiated mature cells to restore function. During recovery, surviving cells activate protective pathways to help epithelial cells regain their properties. This repair involves processes such as dedifferentiation, metabolic changes, extracellular matrix production, and nephron hypertrophy. If successful, these coordinated actions result in adaptive repair, allowing the kidneys to reach a new equilibrium, as seen in the rapid recovery of kidney function in young patients or after mild injury. However, despite an incomplete understanding of the transition from AKI to CKD, current evidence suggests that maladaptive repair—a process where inappropriate cellular reactions lead to improper healing—is the most likely mechanism driving chronic kidney damage[36]. Maladaptive repair in tubular, vascular, and interstitial compartments after AKI can lead to progressive renal interstitial fibrosis. This occurs through mechanisms such as cell cycle arrest, secretion of profibrotic cytokines, pericyte activation leading to myofibroblast generation, inflammation, loss of peritubular capillaries, and increased production of extracellular matrix. Collectively, these processes contribute to long-term damage and dysfunction of the kidneys[37].

After AKI, the activation of the DNA damage response is essential for initiating repair in renal epithelial cells. When complete repair fails, proximal tubules may enter G2/M cell cycle arrest to maintain genomic stability. Although initially protective, prolonged G2/M arrest can promote a profibrotic state, leading to fibrosis and irreversible damage[38,39]. Cellular senescence, associated with repeated injury and DNA damage, accelerates fibrosis and mimics aging[40]. The role of epigenetic modifications is gaining attention lately, as changes in DNA methylation, histone modifications, and noncoding RNAs are believed to play a crucial role. These changes, often triggered by hypoxia, promote the expression of inflammatory genes and increase collagen secretion, contributing to fibrosis[41]. Both innate and adaptive immune responses, such as macrophages, complements, and T cells, are involved in this transition[37,42]. Mitochondrial dysfunction in proximal tubules, including disrupted homeostasis and mtDNA release, triggers immune responses that activate inflammatory pathways. Hypoxia during AKI exacerbates mitochondrial dysfunction, leading to capillary rarefaction and further kidney injury[43,44].

Risk factors and predictors of AKI to CKD transition

The frequent use of diuretics, nephrotoxic medications, and uncontrolled ascites significantly contribute to the persistence of AKI and increase the risk of progression to CKD[6,45]. Additionally, besides HRS-CKD, other liver-associated renal pathologies such as immunoglobulin A nephropathy, hepatitis B virus or hepatitis C virus-associated glomerulonephritis, and polyarteritis nodosa further predispose cirrhosis patients to CKD[46,47]. Systemic comorbidities known to promote renal injury, including hypertensive nephrosclerosis, diabetic nephropathy, obesity/metabolic syndrome-related nephropathy, and unresolved obstructive uropathy, are risk factors that may further contribute to the progression from AKI to CKD. The presence of concurrent diabetes is one of the most common causes of CKD developing in cirrhosis patients[33]. Diabetes mellitus is highly prevalent in cirrhosis, with rates between 35% and 71%, significantly exceeding those in the general population[48]. A recent study by Maji et al[49] found that apart from type 2 diabetes, cirrhosis itself may lead to a form of diabetes known as hepatogenous diabetes, which is the predominant type in child C cirrhosis patients. Another important risk factor for CKD is MASLD. MASLD and CKD are closely linked, primarily due to shared risk factors such as obesity, insulin resistance, and metabolic syndrome. The prevalence of MASLD among CKD patients is significant, with studies indicating that 30%–40% of individuals with CKD also have MASLD[50]. In a meta-analysis by Musso et al[51], the incidence and prevalence of CKD are approximately twice as high in MASLD patients. Thus, screening for kidney function in MASLD patients is crucial for early interventions. In cirrhotic patients with AKI, baseline elevated serum cystatin C levels and higher model for end-stage liver disease scores also predict poor renal recovery. Additionally, higher initial AKI stages—particularly stages 2 and 3—predict a greater likelihood of CKD development[33]. Each new episode of AKI further impairs the kidney's autoregulation mechanisms, ultimately resulting in interstitial fibrosis rather than regeneration[52].

Clinical implications of CKD in cirrhosis

In cirrhosis patients, CKD is associated with significantly worse outcomes[3]. They experience higher rates of AKI (68% vs 21%), an increased need for dialysis (11% vs 2%), and a higher 30-day mortality rate (16% vs 7%) compared to cirrhosis patients without CKD. Further, CKD is also associated with more frequent occurrences of refractory ascites (25% vs 7%), bacterial infections (58% vs 34%), and a greater likelihood of LT (25% vs 10%), as well as higher rates of hospitalization (70% vs 63%) compared to those without CKD[4]. Similarly, Bassegoda et al[6] also reported that patients with cirrhosis and CKD had a greater frequency of AKI (75% vs 45%), refractory ascites (25% vs 7%), bacterial infections (58% vs 34%), and a higher likelihood of needing LT (25% vs 10%) compared to those without CKD[5]. Cullaro et al[35] highlighted that one-year post-LT mortality was also higher in patients with CKD (12% vs 9%), suggesting that CKD may impact renal outcomes after transplantation too.

Patients with cirrhosis and CKD often experience overlapping symptoms such as ascites, edema, and bleeding tendencies, complicating the determination of disease-specific contributions and the selection of appropriate therapies. CKD can exacerbate other manifestations of cirrhosis, including anemia, immunodepression, and bleeding tendencies. Additionally, CKD raises cardiovascular mortality risk in cirrhosis patients[47]. CKD is also linked to a higher prevalence of hepatocellular carcinoma and other cancers, particularly in patients with MASLD. Shared risk factors such as alcohol use and viral hepatitis further contribute to the complexity of managing these patients[53-55]. Standard therapies like hemodialysis are poorly tolerated by cirrhosis patients with CKD compared to peritoneal dialysis[56]. To summarize, up to 25% of cirrhosis patients who recover from AKI may progress to CKD, potentially due to maladaptive repair processes and ongoing hemodynamic alterations. This risk is heightened by concurrent risk factors, such as diabetes and MASLD. Progression to CKD is associated with various complications and poorer clinical outcomes in cirrhosis patients.

EARLY IDENTIFICATION OF AKI TO CKD TRANSITION

To improve renal outcomes in cirrhosis patients, early identification of persistent AKI, AKD, and CKD is crucial. Traditional tools, such as creatinine-based eGFR equations, often overestimate GFR, leading to delayed diagnosis[57]. Cystatin C-based eGFR calculation may be a better tool for assessing kidney function in cirrhosis, as it is less affected by hepatic function, muscle mass, and serum bilirubin, enabling earlier CKD detection[58]. However, cost, availability, and lack of standardization limit its use[59]. Conventional urine indicators such as albuminuria are less accurate in cirrhosis patients due to hypoalbuminemia and increased capillary permeability[60]. These factors underscore the need for novel biomarkers of kidney damage that may be present before the development of functional abnormalities. Kidney injury molecule 1 (KIM-1), a type 1 transmembrane glycoprotein, is expressed in response to mainly tubular injury and is upregulated in dedifferentiated proximal tubular epithelial cells after ischemic or toxic injury. Urinary KIM-1 is a good predictor of renal injury before detectable changes in eGFR, making it a potential biomarker for early detection of CKD[61]. Neutrophil gelatinase-associated lipocalin (NGAL), produced by renal tubular cells in response to injury, is a highly studied biomarker for renal dysfunction in cirrhosis patients. NGAL is a rapid-response biomarker for kidney damage, detectable within two hours of injury[62]. Its role in inflammation and cell proliferation also links it to CKD progression. Elevated levels of NGAL in plasma and urine correlate inversely with eGFR, reflecting its predictive value for renal function decline[63]. Urinary NGAL has shown a specificity of 83.3% and a sensitivity of 70.6% in predicting microalbuminuria and early kidney damage, suggesting its potential to stratify CKD progression risk post-AKI[64,65]. There are multiple point-of-care test devices available for quick measurement of plasma/urine NGAL, providing results within 15–20 minutes. MicroRNAs (miRNAs), non-coding RNAs that regulate gene expression, are also promising biomarkers for CKD. Circulating miR-21, associated with fibrosis, correlates inversely with renal function, while urinary miRNAs are stable and detectable even in early CKD stages[66-68]. Specific miRNA families, such as miR-29 and miR-200, diminish with kidney damage, indicating disease severity[68]. Other potential biomarkers of CKD include uromodulin, which decreases with reduced nephron mass[69], and dimethylarginine, markers of endothelial dysfunction that increase with CKD progression[70]. Lastly, emerging proteomic and metabolomic panels, like the CKD273 urinary peptide panel, may further improve the early detection of CKD[71,72]. Plasma concentrations of soluble tumor necrosis factor receptors 1 and 2 have been consistently found to be associated with incident CKD in nondiabetic subjects[73]. The CKD Biomarkers Consortium's protocols have yielded important insights into the role and validation of biomarkers. Their research found that elevated plasma and urine NGAL were associated with faster eGFR decline and progression to ESRD. NGAL also predicted the onset of albuminuria and structural kidney damage. These markers are particularly useful when combined with traditional risk factors to CKD, like proteinuria and other clinical parameters. Results of phase II protocols found that higher NGAL levels when combined with KIM-1 and IL-18 predicted adverse outcomes including hospitalisation, rapid CKD progression and mortality[74]. In another prospective study by Wen et al[75], involving 656 patients hospitalised with AKI followed up with 7 different biomarkers, each standard deviation increase in KIM-1 from baseline to 12 months, along with other biomarkers, increases risk of progression to CKD by 2-3 times.

The use of biomarkers in the routine clinical practice is generally constrained by cost, availability, and lack of standardization. The current biomarkers of AKI have limited efficacy in predicting AKD or CKD on their own. There is a pressing need for the development of novel biomarkers that can accurately predict the transition from AKI to CKD. The CKD Biomarker Consortium is actively working toward this goal. Besides KIM-1 and NGAL, other markers such as liver fatty acid-binding protein, interleukin-18, and tissue inhibitor of metalloproteinases (TIMP)-2 × insulin-like growth factor-binding protein-7 (IGFBP7) reflect sustained tubular injury, indicating a higher risk of CKD development[76]. A study of 692 patients found that TIMP-2 × IGFBP7 levels above 2.0 at admission were associated with increased mortality or the need for dialysis within nine months[77]. Given the role of the renin-angiotensin-aldosterone system in CKD progression, urinary angiotensinogen could serve as a useful marker[78]. The acute dialysis quality index has recently proposed a staging system for AKD that incorporates traditional markers like albuminuria, hematuria, and imaging findings, along with newer biomarkers to better stratify AKD patients at high risk of progression[79]. Additionally, the Chronic Renal Insufficiency study identified rapid CKD progressors with specific DNA methylation patterns linked to inflammation and fibrosis, particularly involving transforming growth factor (TGF)-β[80].

Fibrosis onset in the kidney is a key indicator of CKD progression. Although tissue biopsy is the gold standard for assessing renal fibrosis, it is invasive, prone to sampling error and observer variability, and can be problematic for patients with advanced cirrhosis. Non-invasive assessment of renal fibrosis has shown promise because of recent advancements in diagnostic imaging techniques including magnetic resonance imaging (MRI) and ultrasound. These imaging techniques quantify renal fibrosis by evaluating its impact on the molecular, mechanical, and functional properties of the kidney, including water mobility restriction by diffusion MRI, tissue hypoxia by blood-oxygenation-level-dependent MRI, renal stiffness by magnetic resonance (MR) and ultrasound elastography. Moreover, other MR techniques like susceptibility-weighted imaging and T1/T2 mapping have been used to quantify renal fibrosis[81-85]. The renal resistive index (RRI), measured by renal duplex ultrasound, is a marker of renal vascular resistance that has potential in assessing functional CKD. RRI can also predict CKD progression as it correlates with renal histopathological changes such as glomerular sclerosis and interstitial fibrosis[86]. Surya et al[87] studied 200 consecutive cirrhosis patients and found RRI could stratified different AKI phenotypes and predicted AKI mortality. As the severity of AKI is a crucial factor for progression, RRI may identify patients at high risk for AKI to CKD progression. In summary, biomarkers of kidney damage can play a crucial role in identifying the transition from AKI to CKD. While several biomarkers have been developed, existing AKI biomarkers face challenges, including limited ability to predict CKD progression, high costs, poor accessibility, and lack of standardization. Further research is necessary to address these gaps. Furthermore, non-invasive imaging techniques for assessing kidney fibrosis have garnered significant attention due to promising results. However, these methods require further refinement to ensure their clinical utility and practicality.

THERAPEUTIC IMPLICATIONS

Addressing AKI promptly in cirrhosis patients and providing appropriate and timely treatment will lead to early recovery from AKI and reduce the progression to CKD. The primary objective in managing HRS-AKI is to reverse the hemodynamic alterations leading to renal vasoconstriction. Management is also tailored to address the underlying cause and precipitating factors of AKI. In HRS-AKI patients, terlipressin plus albumin has been shown to achieve a significantly higher rate of HRS reversal (27% vs 14%, P = 0.004) compared to albumin alone[88]. Terlipressin plus albumin is also more effective in reversing HRS-AKI than a combination of midodrine and octreotide plus albumin[89]. Early institution of therapy results in a better response rate. In a recent study, AKI reversal was higher and earlier in patients with ACLF when terlipressin was administered after 12 hours of volume expansion, rather than 48 hours (68.6% vs 31.4%, P = 0.03)[90]. Reversal of HRS-AKI also reduces the risk of developing CKD after LT. A study demonstrated that patients who responded to terlipressin and albumin had a significantly lower probability of developing CKD one year post-LT compared to non-responders (31% vs 65%, P = 0.019)[91]. It is essential to note that vasoconstrictors are more effective in treating HRS when initiated at lower serum creatinine levels (< 2.25 mg/dL) and when achieving a significant increase in MAP of 15 mmHg or more[3]. Reversal of HRS is less likely in patients with higher baseline creatinine, lower baseline MAP, underlying cirrhotic cardiomyopathy, or portopulmonary hypertension. Transjugular intrahepatic portosystemic shunt (TIPS) placement has been shown to reverse HRS and improve renal and cardiac function by reducing portal hypertension. In a metanalysis of 9 studies, the pooled rate of renal function improvement after TIPS was 83% in any type of HRS, although with a high incidence of hepatic encephalopathy (49%)[92].

Emerging therapies aimed at preventing the transition from AKI to CKD in cirrhotic patients focus on managing inflammation, oxidative stress, and fibrosis, as well as promoting kidney repair and regeneration. The role of antifibrotic therapy is still evolving and is currently hindered by poor specificity, incomplete fibrosis reversal, off-target effects, and limited clinical trials validating long-term outcomes. Targeted antifibrotic therapies, such as those inhibiting TGF-β signalling, are under investigation to reduce kidney fibrosis and inflammation—key contributors to CKD progression[93,94]. Pirfenidone, an antifibrotic drug, inhibits the expression of TGF-β and exhibits anti-inflammatory activity by suppressing tumor necrosis factor expression. Approved for idiopathic pulmonary fibrosis in 2021, it is currently under investigation for renal fibrosis[95,96]. In a double-blind randomized controlled trial involving 77 participants with diabetic nephropathy, the pirfenidone 1200 mg/day group showed a mean eGFR increase of 3.3 mL/minute/1.73 m² ± 8.5 mL/minute/1.73 m²[97]. The results of the ongoing Phase 2 TOP-CKD trial (ID: NCT04258397), the largest trial on pirfenidone for kidney fibrosis using imaging and urinary biomarkers, are awaited. Another drug, Lademirsen, is a microRNA inhibitor targeting miR-21, which regulates kidney fibrosis. A Phase 2 trial of Lademirsen is underway for Alport's syndrome (HERA; trial ID: NCT02855268)[96]. Hypoxia-inducible factor stabilizers, such as FG-4592 (Roxadustat), have shown promising effects by supporting renal vascular regeneration and reducing oxidative stress. This helps alleviate ischemia-induced renal injury and can decrease the likelihood of fibrosis and long-term CKD development[98]. Similarly, therapies aimed at modulating the renin-angiotensin-aldosterone system have shown potential for slowing fibrosis and improving renal outcomes in cirrhotic AKI patients[99].

Newer interventions targeting endothelial dysfunction and oxidative stress, such as the use of nitric oxide donors and antioxidants, may also offer protective effects for these patients by improving blood flow to the kidneys[100]. Mesenchymal stem cell (MSC) therapies have demonstrated potential in reducing renal fibrosis and encouraging kidney repair by releasing anti-inflammatory exosomes[101]. Currently, the idea of using stem cell therapy for renal fibrosis is based on inhibiting signalling pathways involved in the development of fibrosis, including both TGF-β/Smad-dependent and independent pathways. Most studies involving stem cell therapies are in the preclinical phase. Results have shown no significant adverse events, but their efficacy remains controversial[102]. Lang et al[103] demonstrated that bone marrow-derived MSCs could significantly reduce renal fibrosis in a diabetic nephropathy rat model, potentially by inhibiting the TGF-β1/Smad3 signalling pathway. Nevertheless, stem cell therapy in CKD faces limitations such as high costs, immune rejection, ethical concerns, and inconsistent efficacy. Additionally, antioxidant and anti-inflammatory agents, such as lactoferrin and poricoic acid, have shown efficacy in animal models[104,105]. In combination with melatonin, these agents help reduce oxidative stress and inflammation, attenuating fibrosis, and the transition from AKI to CKD. Particularly, Lactoferrin enhances kidney autophagy while inhibiting apoptosis, promoting a more resilient renal environment less prone to fibrosis[104]. The emerging field of nanotherapeutics is being investigated as a novel treatment strategy for AKI. Recent developments have led to the creation of various nanomaterials, including molybdenum-based polyoxometalate nanoclusters, mitochondria-targeting ceria nanoparticles, and black phosphorus nanosheets. These nanomaterials have been shown to scavenge reactive oxygen species and reduce inflammation in various AKI models[106]. Biomarker-driven therapies are another promising avenue guiding individualized treatment plans[107,108]. Thus, our growing understanding of kidney injury and repair mechanisms is facilitating the creation of targeted treatments, currently being tested in clinical trials. As precision medicine advances, these targeted therapies and early interventions hold promise for reducing the transition from AKI to CKD.

CONCLUSION

The transition from AKI to CKD in cirrhotic patients poses a significant clinical challenge, underscoring the urgent need for early detection, prompt intervention, and comprehensive management strategies. Table 1 summarizes the critical issues involving the transition from AKI to CKD in patients with liver cirrhosis. Clinicians should prioritize the utilization of novel biomarkers and the development of non-invasive imaging techniques, such as renal elastography and MRI-based fibrosis assessment, to enhance monitoring and inform therapeutic decisions. Future research should aim to refine these biomarkers, improving diagnostic accuracy in the AKI-to-CKD continuum, particularly in the unique context of cirrhosis. Additionally, innovative therapeutic strategies targeting maladaptive repair mechanisms, including cellular senescence, fibrotic signalling, and oxidative damage, may offer avenues to mitigate or even prevent CKD progression. Ultimately, a multidisciplinary approach that integrates early detection, advanced monitoring, and targeted interventions is crucial for improving renal outcomes and enhancing the quality of life for cirrhosis patients at risk for CKD.

Table 1 Important issues involving transition of acute kidney injury to chronic kidney disease in patients with liver cirrhosis.
Factors
Description
Risk factors for CKD transitionAKI characteristics: Recurrent AKI, severe AKI, acute kidney disease
Metabolic factors: Diabetes mellitus, hypertension, metabolic dysfunction-associated steatotic liver disease
Cirrhosis factors: Higher model for end-stage liver disease scores, neuro-hormonal alterations
Etiological factors: Alcohol, hepatis B virus and hepatitis C virus (glomerulonephritis)
Putative mechanisms for chronic liver disease transitionMaladaptive repair mechanism: Cell cycle arrest and cellular senescence
Inflammation and fibrosis: Profibrotic cytokines, myofibroblast generation, and increase in extracellular matrix
Clinical impacts of CKDRefractory ascites and increased frequency of bacterial infection
Increased frequency of AKI
Need for dialysis and need for liver transplantation and simultaneous liver and kidney transplantation
Higher mortality risk
Novel biomarkers for AKI-CKDKidney injury molecule-1
Neutrophil gelatinase-associated lipocalin
Tissue inhibitor of metalloproteinases-2
Uromodulin and dimethylarginine
Liver type-fatty acid binding protein
Future potential therapiesTargeted transforming growth factor-β inhibition
Modulation of renin-angiotensin-aldosterone system, endothelial dysfunction and oxidative stress biomarker driven therapy
Hypoxia inducible factor stabilizers
Mesenchymal stem cell therapies
Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Urology and nephrology

Country of origin: India

Peer-review report’s classification

Scientific Quality: Grade A, Grade B, Grade C, Grade E

Novelty: Grade B, Grade B, Grade C, Grade C

Creativity or Innovation: Grade A, Grade C, Grade C, Grade C

Scientific Significance: Grade B, Grade B, Grade B, Grade C

P-Reviewer: Huang YX; Shrestha B; Zhang JQ S-Editor: Luo ML L-Editor: A P-Editor: Zhao S

References
1.  Nadim MK, Garcia-Tsao G. Acute Kidney Injury in Patients with Cirrhosis. N Engl J Med. 2023;388:733-745.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 39]  [Reference Citation Analysis (0)]
2.  D'hoore E, Neirynck N, Schepers E, Vanholder R, Verbeke F, Van Thielen M, Van Biesen W. Chronic kidney disease progression is mainly associated with non-recovery of acute kidney injury. J Nephrol. 2015;28:709-716.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
3.  Nadim MK, Kellum JA, Forni L, Francoz C, Asrani SK, Ostermann M, Allegretti AS, Neyra JA, Olson JC, Piano S, VanWagner LB, Verna EC, Akcan-Arikan A, Angeli P, Belcher JM, Biggins SW, Deep A, Garcia-Tsao G, Genyk YS, Gines P, Kamath PS, Kane-Gill SL, Kaushik M, Lumlertgul N, Macedo E, Maiwall R, Marciano S, Pichler RH, Ronco C, Tandon P, Velez JQ, Mehta RL, Durand F. Acute kidney injury in patients with cirrhosis: Acute Disease Quality Initiative (ADQI) and International Club of Ascites (ICA) joint multidisciplinary consensus meeting. J Hepatol. 2024;81:163-183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Reference Citation Analysis (0)]
4.  Wong F, Reddy KR, O'Leary JG, Tandon P, Biggins SW, Garcia-Tsao G, Maliakkal BJ, Lai JC, Fallon MB, Vargas HE, Subramanian R, Thuluvath PJ, Kamath PS, Thacker L, Bajaj JS. Impact of Chronic Kidney Disease on Outcomes in Cirrhosis. Liver Transpl. 2019;25:870-880.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 43]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
5.  Younossi ZM, Golabi P, Paik JM, Henry A, Van Dongen C, Henry L. The global epidemiology of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH): a systematic review. Hepatology. 2023;77:1335-1347.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 270]  [Cited by in F6Publishing: 894]  [Article Influence: 447.0]  [Reference Citation Analysis (2)]
6.  Bassegoda O, Huelin P, Ariza X, Solé C, Juanola A, Gratacós-Ginès J, Carol M, Graupera I, Pose E, Napoleone L, Albertos S, de Prada G, Cervera M, Fernández J, Fabrellas N, Poch E, Solà E, Ginès P. Development of chronic kidney disease after acute kidney injury in patients with cirrhosis is common and impairs clinical outcomes. J Hepatol. 2020;72:1132-1139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 39]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
7.  Tariq R, Hadi Y, Chahal K, Reddy S, Salameh H, Singal AK. Incidence, Mortality and Predictors of Acute Kidney Injury in Patients with Cirrhosis: A Systematic Review and Meta-analysis. J Clin Transl Hepatol. 2020;8:135-142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 27]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
8.  Jiang W, Hu Y, Sun Y, Shen Y, Xun Y. Prevalence and short-term outcome of acute kidney injury in patients with acute-on-chronic liver failure: A meta-analysis. J Viral Hepat. 2020;27:810-817.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
9.  Bai Z, Primignani M, Guo X, Zheng K, Li H, Qi X. Incidence and mortality of renal dysfunction in cirrhotic patients with acute gastrointestinal bleeding: a systematic review and meta-analysis. Expert Rev Gastroenterol Hepatol. 2019;13:1181-1188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 15]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
10.  Kellum JA, Lameire N; KDIGO AKI Guideline Work Group. Diagnosis, evaluation, and management of acute kidney injury: a KDIGO summary (Part 1). Crit Care. 2013;17:204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1192]  [Cited by in F6Publishing: 1696]  [Article Influence: 141.3]  [Reference Citation Analysis (0)]
11.  Lekakis V, Gkoufa A, Vlachogiannakos J, Papatheodoridis GV, Cholongitas E. Incidence and risk factors of acute kidney injury in cirrhosis: a systematic review and meta-analysis of 5,202,232 outpatients, inpatients, and ICU-admitted patients. Expert Rev Gastroenterol Hepatol. 2024;18:377-388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
12.  Velez JCQ, Therapondos G, Juncos LA. Reappraising the spectrum of AKI and hepatorenal syndrome in patients with cirrhosis. Nat Rev Nephrol. 2020;16:137-155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 75]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
13.  Coca SG, Singanamala S, Parikh CR. Chronic kidney disease after acute kidney injury: a systematic review and meta-analysis. Kidney Int. 2012;81:442-448.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1265]  [Cited by in F6Publishing: 1496]  [Article Influence: 106.9]  [Reference Citation Analysis (0)]
14.  Heung M, Steffick DE, Zivin K, Gillespie BW, Banerjee T, Hsu CY, Powe NR, Pavkov ME, Williams DE, Saran R, Shahinian VB; Centers for Disease Control and Prevention CKD Surveillance Team. Acute Kidney Injury Recovery Pattern and Subsequent Risk of CKD: An Analysis of Veterans Health Administration Data. Am J Kidney Dis. 2016;67:742-752.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 221]  [Cited by in F6Publishing: 263]  [Article Influence: 26.3]  [Reference Citation Analysis (0)]
15.  Patidar KR, Cullaro G, Naved MA, Kabir S, Grama A, Orman ES, Piano S, Allegretti AS. Prognostic significance of acute kidney injury stage 1B in hospitalized patients with cirrhosis: A US nationwide study. Liver Transpl. 2024;30:244-253.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
16.  Patidar KR, Naved MA, Kabir S, Grama A, Allegretti AS, Cullaro G, Asrani SK, Worden A, Desai AP, Ghabril MS, Nephew LD, Orman ES. Longer time to recovery from acute kidney injury is associated with major adverse kidney events in patients with cirrhosis. Aliment Pharmacol Ther. 2023;57:1397-1406.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
17.  Worden A, Pike F, Allegretti AS, Kaur H, Peng JL, Kettler CD, Orman ES, Desai AP, Nephew LD, Ghabril MS, Patidar KR. The prognostic impact of acute kidney injury recovery patterns in critically ill patients with cirrhosis. Liver Transpl. 2023;29:246-258.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
18.  Levey AS. Defining AKD: The Spectrum of AKI, AKD, and CKD. Nephron. 2022;146:302-305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 67]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
19.  Chawla LS, Bellomo R, Bihorac A, Goldstein SL, Siew ED, Bagshaw SM, Bittleman D, Cruz D, Endre Z, Fitzgerald RL, Forni L, Kane-Gill SL, Hoste E, Koyner J, Liu KD, Macedo E, Mehta R, Murray P, Nadim M, Ostermann M, Palevsky PM, Pannu N, Rosner M, Wald R, Zarbock A, Ronco C, Kellum JA; Acute Disease Quality Initiative Workgroup 16. Acute kidney disease and renal recovery: consensus report of the Acute Disease Quality Initiative (ADQI) 16 Workgroup. Nat Rev Nephrol. 2017;13:241-257.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 645]  [Cited by in F6Publishing: 956]  [Article Influence: 119.5]  [Reference Citation Analysis (0)]
20.  Chawla LS, Eggers PW, Star RA, Kimmel PL. Acute kidney injury and chronic kidney disease as interconnected syndromes. N Engl J Med. 2014;371:58-66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1210]  [Cited by in F6Publishing: 1383]  [Article Influence: 125.7]  [Reference Citation Analysis (0)]
21.  Pannu N. Bidirectional relationships between acute kidney injury and chronic kidney disease. Curr Opin Nephrol Hypertens. 2013;22:351-356.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 31]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
22.  Lameire NH, Levin A, Kellum JA, Cheung M, Jadoul M, Winkelmayer WC, Stevens PE; Conference Participants. Harmonizing acute and chronic kidney disease definition and classification: report of a Kidney Disease: Improving Global Outcomes (KDIGO) Consensus Conference. Kidney Int. 2021;100:516-526.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 217]  [Article Influence: 54.3]  [Reference Citation Analysis (0)]
23.  Khwaja A. KDIGO clinical practice guidelines for acute kidney injury. Nephron Clin Pract. 2012;120:c179-c184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1436]  [Cited by in F6Publishing: 2949]  [Article Influence: 226.8]  [Reference Citation Analysis (0)]
24.  Angeli P, Garcia-Tsao G, Nadim MK, Parikh CR. News in pathophysiology, definition and classification of hepatorenal syndrome: A step beyond the International Club of Ascites (ICA) consensus document. J Hepatol. 2019;71:811-822.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 245]  [Article Influence: 40.8]  [Reference Citation Analysis (0)]
25.  Tonon M, Rosi S, Gambino CG, Piano S, Calvino V, Romano A, Martini A, Pontisso P, Angeli P. Natural history of acute kidney disease in patients with cirrhosis. J Hepatol. 2021;74:578-583.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 29]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
26.  James MT, Levey AS, Tonelli M, Tan Z, Barry R, Pannu N, Ravani P, Klarenbach SW, Manns BJ, Hemmelgarn BR. Incidence and Prognosis of Acute Kidney Diseases and Disorders Using an Integrated Approach to Laboratory Measurements in a Universal Health Care System. JAMA Netw Open. 2019;2:e191795.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 77]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
27.  See EJ, Polkinghorne KR, Toussaint ND, Bailey M, Johnson DW, Bellomo R. Epidemiology and Outcomes of Acute Kidney Diseases: A Comparative Analysis. Am J Nephrol. 2021;52:342-350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
28.  Orieux A, Prezelin-Reydit M, Prevel R, Combe C, Gruson D, Boyer A, Rubin S. Clinical trajectories and impact of acute kidney disease after acute kidney injury in the intensive care unit: a 5-year single-centre cohort study. Nephrol Dial Transplant. 2023;38:167-176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
29.  Vanmassenhove J, Vanholder R, Lameire N. Points of Concern in Post Acute Kidney Injury Management. Nephron. 2018;138:92-103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 20]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
30.  Wong F, Nadim MK, Kellum JA, Salerno F, Bellomo R, Gerbes A, Angeli P, Moreau R, Davenport A, Jalan R, Ronco C, Genyk Y, Arroyo V. Working Party proposal for a revised classification system of renal dysfunction in patients with cirrhosis. Gut. 2011;60:702-709.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 295]  [Cited by in F6Publishing: 278]  [Article Influence: 19.9]  [Reference Citation Analysis (0)]
31.  Angeli P, Ginès P, Wong F, Bernardi M, Boyer TD, Gerbes A, Moreau R, Jalan R, Sarin SK, Piano S, Moore K, Lee SS, Durand F, Salerno F, Caraceni P, Kim WR, Arroyo V, Garcia-Tsao G. Diagnosis and management of acute kidney injury in patients with cirrhosis: revised consensus recommendations of the International Club of Ascites. J Hepatol. 2015;62:968-974.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 432]  [Cited by in F6Publishing: 498]  [Article Influence: 49.8]  [Reference Citation Analysis (1)]
32.  Solà E, Ginès P. Chronic Kidney Disease in Cirrhosis: Emerging Complication With Negative Impact in the Liver Transplant Setting. Liver Transpl. 2020;26:483-484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
33.  Maiwall R, Pasupuleti SSR, Bihari C, Rastogi A, Singh PK, Naik V, Singh A, Jain P, Kumar A, Mukund A, Mathur RP, Kumar G, Sarin SK. Incidence, Risk Factors, and Outcomes of Transition of Acute Kidney Injury to Chronic Kidney Disease in Cirrhosis: A Prospective Cohort Study. Hepatology. 2020;71:1009-1022.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 54]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
34.  Chen CY, Lin CJ, Lin CS, Sun FJ, Pan CF, Chen HH, Wu CJ. The prevalence and association of chronic kidney disease and diabetes in liver cirrhosis using different estimated glomerular filtration rate equation. Oncotarget. 2018;9:2236-2248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
35.  Cullaro G, Verna EC, Lee BP, Lai JC. Chronic Kidney Disease in Liver Transplant Candidates: A Rising Burden Impacting Post-Liver Transplant Outcomes. Liver Transpl. 2020;26:498-506.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 40]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
36.  Rayego-Mateos S, Marquez-Expósito L, Rodrigues-Diez R, Sanz AB, Guiteras R, Doladé N, Rubio-Soto I, Manonelles A, Codina S, Ortiz A, Cruzado JM, Ruiz-Ortega M, Sola A. Molecular Mechanisms of Kidney Injury and Repair. Int J Mol Sci. 2022;23:1542.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 31]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
37.  Yu SM, Bonventre JV. Acute kidney injury and maladaptive tubular repair leading to renal fibrosis. Curr Opin Nephrol Hypertens. 2020;29:310-318.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 84]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
38.  Zhang Y, Yang Y, Yang F, Liu X, Zhan P, Wu J, Wang X, Wang Z, Tang W, Sun Y, Zhang Y, Xu Q, Shang J, Zhen J, Liu M, Yi F. HDAC9-mediated epithelial cell cycle arrest in G2/M contributes to kidney fibrosis in male mice. Nat Commun. 2023;14:3007.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 9]  [Reference Citation Analysis (0)]
39.  Canaud G, Bonventre JV. Cell cycle arrest and the evolution of chronic kidney disease from acute kidney injury. Nephrol Dial Transplant. 2015;30:575-583.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 149]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
40.  Ferenbach DA, Bonventre JV. Mechanisms of maladaptive repair after AKI leading to accelerated kidney ageing and CKD. Nat Rev Nephrol. 2015;11:264-276.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 430]  [Cited by in F6Publishing: 567]  [Article Influence: 56.7]  [Reference Citation Analysis (0)]
41.  Li Z, Li N. Epigenetic Modification Drives Acute Kidney Injury-to-Chronic Kidney Disease Progression. Nephron. 2021;145:737-747.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
42.  Liu J, Kumar S, Dolzhenko E, Alvarado GF, Guo J, Lu C, Chen Y, Li M, Dessing MC, Parvez RK, Cippà PE, Krautzberger AM, Saribekyan G, Smith AD, McMahon AP. Molecular characterization of the transition from acute to chronic kidney injury following ischemia/reperfusion. JCI Insight. 2017;2:e94716.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 200]  [Article Influence: 25.0]  [Reference Citation Analysis (0)]
43.  Chung KW, Dhillon P, Huang S, Sheng X, Shrestha R, Qiu C, Kaufman BA, Park J, Pei L, Baur J, Palmer M, Susztak K. Mitochondrial Damage and Activation of the STING Pathway Lead to Renal Inflammation and Fibrosis. Cell Metab. 2019;30:784-799.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 322]  [Cited by in F6Publishing: 372]  [Article Influence: 62.0]  [Reference Citation Analysis (0)]
44.  Koh ES, Chung S. Recent Update on Acute Kidney Injury-to-Chronic Kidney Disease Transition. Yonsei Med J. 2024;65:247-256.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
45.  Bucsics T, Krones E. Renal dysfunction in cirrhosis: acute kidney injury and the hepatorenal syndrome. Gastroenterol Rep (Oxf). 2017;5:127-137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 59]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
46.  Venkataseshan VS, Lieberman K, Kim DU, Thung SN, Dikman S, D'Agati V, Susin M, Valderrama E, Gauthier B, Prakash A. Hepatitis-B-associated glomerulonephritis: pathology, pathogenesis, and clinical course. Medicine (Baltimore). 1990;69:200-216.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Kumar R, Priyadarshi RN, Anand U. Chronic renal dysfunction in cirrhosis: A new frontier in hepatology. World J Gastroenterol. 2021;27:990-1005.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 8]  [Cited by in F6Publishing: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (2)]
48.  García-Compeán D, Orsi E, Kumar R, Gundling F, Nishida T, Villarreal-Pérez JZ, Del Cueto-Aguilera ÁN, González-González JA, Pugliese G. Clinical implications of diabetes in chronic liver disease: Diagnosis, outcomes and management, current and future perspectives. World J Gastroenterol. 2022;28:775-793.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 13]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (7)]
49.  Maji T, Mahto M, Kumar S, Anand U, Priyadarshi RN, Arya R, Kumar R. Hepatogenous Diabetes as Compared to Type-2 Diabetes Mellitus and Non-diabetes in Patients With Liver Cirrhosis: Magnitude, Characteristics, and Implications. J Clin Exp Hepatol. 2024;14:101411.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
50.  Sinn DH, Kang D, Jang HR, Gu S, Cho SJ, Paik SW, Ryu S, Chang Y, Lazo M, Guallar E, Cho J, Gwak GY. Development of chronic kidney disease in patients with non-alcoholic fatty liver disease: A cohort study. J Hepatol. 2017;67:1274-1280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 106]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
51.  Musso G, Gambino R, Tabibian JH, Ekstedt M, Kechagias S, Hamaguchi M, Hultcrantz R, Hagström H, Yoon SK, Charatcharoenwitthaya P, George J, Barrera F, Hafliðadóttir S, Björnsson ES, Armstrong MJ, Hopkins LJ, Gao X, Francque S, Verrijken A, Yilmaz Y, Lindor KD, Charlton M, Haring R, Lerch MM, Rettig R, Völzke H, Ryu S, Li G, Wong LL, Machado M, Cortez-Pinto H, Yasui K, Cassader M. Association of non-alcoholic fatty liver disease with chronic kidney disease: a systematic review and meta-analysis. PLoS Med. 2014;11:e1001680.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 408]  [Cited by in F6Publishing: 470]  [Article Influence: 42.7]  [Reference Citation Analysis (0)]
52.  Sharma A, Mucino MJ, Ronco C. Renal functional reserve and renal recovery after acute kidney injury. Nephron Clin Pract. 2014;127:94-100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 185]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
53.  Martín-Llahí M, Guevara M, Torre A, Fagundes C, Restuccia T, Gilabert R, Solá E, Pereira G, Marinelli M, Pavesi M, Fernández J, Rodés J, Arroyo V, Ginès P. Prognostic importance of the cause of renal failure in patients with cirrhosis. Gastroenterology. 2011;140:488-496.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 243]  [Cited by in F6Publishing: 231]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
54.  Chou CL, Chiu HW, Hsu YH, Yu SM, Liou TH, Sung LC. Impact of chronic kidney disease and end-stage renal disease on the mid-term adverse outcomes in diabetic patients with cardiovascular diseases. Sci Rep. 2024;14:15770.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
55.  Tendulkar KK, Cope B, Dong J, Plumb TJ, Campbell WS, Ganti AK. Risk of malignancy in patients with chronic kidney disease. PLoS One. 2022;17:e0272910.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
56.  Chaudhary K, Khanna R. Renal replacement therapy in end-stage renal disease patients with chronic liver disease and ascites: role of peritoneal dialysis. Perit Dial Int. 2008;28:113-117.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Skluzacek PA, Szewc RG, Nolan CR 3rd, Riley DJ, Lee S, Pergola PE. Prediction of GFR in liver transplant candidates. Am J Kidney Dis. 2003;42:1169-1176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 99]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
58.  Mindikoglu AL, Dowling TC, Weir MR, Seliger SL, Christenson RH, Magder LS. Performance of chronic kidney disease epidemiology collaboration creatinine-cystatin C equation for estimating kidney function in cirrhosis. Hepatology. 2014;59:1532-1542.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 94]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
59.  Stevens LA, Schmid CH, Greene T, Li L, Beck GJ, Joffe MM, Froissart M, Kusek JW, Zhang YL, Coresh J, Levey AS. Factors other than glomerular filtration rate affect serum cystatin C levels. Kidney Int. 2009;75:652-660.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 462]  [Cited by in F6Publishing: 536]  [Article Influence: 31.5]  [Reference Citation Analysis (0)]
60.  Francoz C, Nadim MK, Durand F. Kidney biomarkers in cirrhosis. J Hepatol. 2016;65:809-824.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 104]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
61.  Geng J, Qiu Y, Qin Z, Su B. The value of kidney injury molecule 1 in predicting acute kidney injury in adult patients: a systematic review and Bayesian meta-analysis. J Transl Med. 2021;19:105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 37]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
62.  Devarajan P. Neutrophil gelatinase-associated lipocalin: a promising biomarker for human acute kidney injury. Biomark Med. 2010;4:265-280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 218]  [Cited by in F6Publishing: 216]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
63.  Mitsnefes MM, Kathman TS, Mishra J, Kartal J, Khoury PR, Nickolas TL, Barasch J, Devarajan P. Serum neutrophil gelatinase-associated lipocalin as a marker of renal function in children with chronic kidney disease. Pediatr Nephrol. 2007;22:101-108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 174]  [Cited by in F6Publishing: 181]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
64.  Schinstock CA, Semret MH, Wagner SJ, Borland TM, Bryant SC, Kashani KB, Larson TS, Lieske JC. Urinalysis is more specific and urinary neutrophil gelatinase-associated lipocalin is more sensitive for early detection of acute kidney injury. Nephrol Dial Transplant. 2013;28:1175-1185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 60]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
65.  Romejko K, Markowska M, Niemczyk S. The Review of Current Knowledge on Neutrophil Gelatinase-Associated Lipocalin (NGAL). Int J Mol Sci. 2023;24:10470.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 52]  [Reference Citation Analysis (0)]
66.  Mahtal N, Lenoir O, Tinel C, Anglicheau D, Tharaux PL. MicroRNAs in kidney injury and disease. Nat Rev Nephrol. 2022;18:643-662.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 78]  [Reference Citation Analysis (0)]
67.  Glowacki F, Savary G, Gnemmi V, Buob D, Van der Hauwaert C, Lo-Guidice JM, Bouyé S, Hazzan M, Pottier N, Perrais M, Aubert S, Cauffiez C. Increased circulating miR-21 levels are associated with kidney fibrosis. PLoS One. 2013;8:e58014.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 160]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
68.  Nassirpour R, Raj D, Townsend R, Argyropoulos C. MicroRNA biomarkers in clinical renal disease: from diabetic nephropathy renal transplantation and beyond. Food Chem Toxicol. 2016;98:73-88.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 23]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
69.  Prajczer S, Heidenreich U, Pfaller W, Kotanko P, Lhotta K, Jennings P. Evidence for a role of uromodulin in chronic kidney disease progression. Nephrol Dial Transplant. 2010;25:1896-1903.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 93]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
70.  Kielstein JT, Salpeter SR, Bode-Boeger SM, Cooke JP, Fliser D. Symmetric dimethylarginine (SDMA) as endogenous marker of renal function--a meta-analysis. Nephrol Dial Transplant. 2006;21:2446-2451.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 256]  [Cited by in F6Publishing: 280]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
71.  Good DM, Zürbig P, Argilés A, Bauer HW, Behrens G, Coon JJ, Dakna M, Decramer S, Delles C, Dominiczak AF, Ehrich JH, Eitner F, Fliser D, Frommberger M, Ganser A, Girolami MA, Golovko I, Gwinner W, Haubitz M, Herget-Rosenthal S, Jankowski J, Jahn H, Jerums G, Julian BA, Kellmann M, Kliem V, Kolch W, Krolewski AS, Luppi M, Massy Z, Melter M, Neusüss C, Novak J, Peter K, Rossing K, Rupprecht H, Schanstra JP, Schiffer E, Stolzenburg JU, Tarnow L, Theodorescu D, Thongboonkerd V, Vanholder R, Weissinger EM, Mischak H, Schmitt-Kopplin P. Naturally occurring human urinary peptides for use in diagnosis of chronic kidney disease. Mol Cell Proteomics. 2010;9:2424-2437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 356]  [Cited by in F6Publishing: 398]  [Article Influence: 26.5]  [Reference Citation Analysis (0)]
72.  Schlosser P, Surapaneni AL, Borisov O, Schmidt IM, Zhou L, Anderson A, Deo R, Dubin R, Ganz P, He J, Kimmel PL, Li H, Nelson RG, Porter AC, Rahman M, Rincon-Choles H, Shah V, Unruh ML, Vasan RS, Zheng Z, Feldman HI, Waikar SS, Köttgen A, Rhee EP, Coresh J, Grams ME; Chronic Renal Insufficiency Cohort (CRIC) Study Investigators and the CKD Biomarkers Consortium. Association of Integrated Proteomic and Metabolomic Modules with Risk of Kidney Disease Progression. J Am Soc Nephrol. 2024;35:923-935.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Reference Citation Analysis (0)]
73.  Sarnak MJ, Katz R, Ix JH, Kimmel PL, Bonventre JV, Schelling J, Cushman M, Vasan RS, Waikar SS, Greenberg JH, Parikh CR, Coca SG, Sabbisetti V, Jogalekar MP, Rebholz C, Zheng Z, Gutierrez OM, Shlipak MG. Plasma Biomarkers as Risk Factors for Incident CKD. Kidney Int Rep. 2022;7:1493-1501.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 8]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
74.  Braden GL, Landry DL. The Next Frontier: Biomarkers and Artificial Intelligence Predicting Cardiorenal Outcomes in Diabetic Kidney Disease. Kidney360. 2022;3:1480-1483.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Reference Citation Analysis (0)]
75.  Wen Y, Xu L, Melchinger I, Thiessen-Philbrook H, Moledina DG, Coca SG, Hsu CY, Go AS, Liu KD, Siew ED, Ikizler TA, Chinchilli VM, Kaufman JS, Kimmel PL, Himmelfarb J, Cantley LG, Parikh CR; ASSESS-AKI Consortium. Longitudinal biomarkers and kidney disease progression after acute kidney injury. JCI Insight. 2023;8:e167731.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 7]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
76.  Malhotra R, Siew ED. Biomarkers for the Early Detection and Prognosis of Acute Kidney Injury. Clin J Am Soc Nephrol. 2017;12:149-173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 116]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
77.  Koyner JL, Shaw AD, Chawla LS, Hoste EA, Bihorac A, Kashani K, Haase M, Shi J, Kellum JA; Sapphire Investigators. Tissue Inhibitor Metalloproteinase-2 (TIMP-2)∙IGF-Binding Protein-7 (IGFBP7) Levels Are Associated with Adverse Long-Term Outcomes in Patients with AKI. J Am Soc Nephrol. 2015;26:1747-1754.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 166]  [Article Influence: 15.1]  [Reference Citation Analysis (0)]
78.  Yoon SY, Kim JS, Jeong KH, Kim SK. Acute Kidney Injury: Biomarker-Guided Diagnosis and Management. Medicina (Kaunas). 2022;58:340.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 15]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
79.  Ostermann M, Zarbock A, Goldstein S, Kashani K, Macedo E, Murugan R, Bell M, Forni L, Guzzi L, Joannidis M, Kane-Gill SL, Legrand M, Mehta R, Murray PT, Pickkers P, Plebani M, Prowle J, Ricci Z, Rimmelé T, Rosner M, Shaw AD, Kellum JA, Ronco C. Recommendations on Acute Kidney Injury Biomarkers From the Acute Disease Quality Initiative Consensus Conference: A Consensus Statement. JAMA Netw Open. 2020;3:e2019209.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 192]  [Cited by in F6Publishing: 356]  [Article Influence: 71.2]  [Reference Citation Analysis (0)]
80.  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. 2014;29:864-872.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 108]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
81.  Jiang K, Ferguson CM, Lerman LO. Noninvasive assessment of renal fibrosis by magnetic resonance imaging and ultrasound techniques. Transl Res. 2019;209:105-120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 61]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
82.  Yuan H, Huang Q, Wen J, Gao Y. Ultrasound viscoelastic imaging in the noninvasive quantitative assessment of chronic kidney disease. Ren Fail. 2024;46:2407882.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
83.  Inoue T, Kozawa E, Okada H, Inukai K, Watanabe S, Kikuta T, Watanabe Y, Takenaka T, Katayama S, Tanaka J, Suzuki H. Noninvasive evaluation of kidney hypoxia and fibrosis using magnetic resonance imaging. J Am Soc Nephrol. 2011;22:1429-1434.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 243]  [Cited by in F6Publishing: 281]  [Article Influence: 20.1]  [Reference Citation Analysis (0)]
84.  Korsmo MJ, Ebrahimi B, Eirin A, Woollard JR, Krier JD, Crane JA, Warner L, Glaser K, Grimm R, Ehman RL, Lerman LO. Magnetic resonance elastography noninvasively detects in vivo renal medullary fibrosis secondary to swine renal artery stenosis. Invest Radiol. 2013;48:61-68.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 59]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
85.  Grenier N, Gennisson JL, Cornelis F, Le Bras Y, Couzi L. Renal ultrasound elastography. Diagn Interv Imaging. 2013;94:545-550.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 93]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
86.  Spatola L, Andrulli S. Doppler ultrasound in kidney diseases: a key parameter in clinical long-term follow-up. J Ultrasound. 2016;19:243-250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 28]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
87.  Surya H, Kumar R, Priyadarshi RN, Surya Prakash S, Kumar S. Renal resistive index measurements by ultrasound in patients with liver cirrhosis: Magnitude and associations with renal dysfunction. World J Radiol. 2024;16:221-231.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (1)]
88.  Sanyal AJ, Boyer TD, Frederick RT, Wong F, Rossaro L, Araya V, Vargas HE, Reddy KR, Pappas SC, Teuber P, Escalante S, Jamil K. Reversal of hepatorenal syndrome type 1 with terlipressin plus albumin vs. placebo plus albumin in a pooled analysis of the OT-0401 and REVERSE randomised clinical studies. Aliment Pharmacol Ther. 2017;45:1390-1402.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 78]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
89.  Gonzalez SA, Chirikov VV, Wang WJ, Huang X, Jamil K, Simonetto DA. Terlipressin vs Midodrine Plus Octreotide for Hepatorenal Syndrome-Acute Kidney Injury: A Propensity Score-Matched Comparison. Clin Transl Gastroenterol. 2023;14:e00627.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
90.  Jindal A, Singh H, Kumar G, Arora V, Sharma MK, Maiwall R, Rajan V, Tewathia HV, Vasishtha C, Sarin SK. Early Versus Standard Initiation of Terlipressin for Acute Kidney Injury in ACLF: A Randomized Controlled Trial (eTerli Study). Dig Dis Sci. 2024;69:2204-2214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
91.  Piano S, Gambino C, Vettore E, Calvino V, Tonon M, Boccagni P, Gringeri E, Germani G, Burra P, Cillo U, Angeli P. Response to Terlipressin and Albumin Is Associated With Improved Liver Transplant Outcomes in Patients With Hepatorenal Syndrome. Hepatology. 2021;73:1909-1919.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 52]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
92.  Song T, Rössle M, He F, Liu F, Guo X, Qi X. Transjugular intrahepatic portosystemic shunt for hepatorenal syndrome: A systematic review and meta-analysis. Dig Liver Dis. 2018;50:323-330.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 58]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
93.  Isaka Y. Targeting TGF-β Signaling in Kidney Fibrosis. Int J Mol Sci. 2018;19:2532.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 170]  [Article Influence: 24.3]  [Reference Citation Analysis (0)]
94.  Akhurst RJ, Hata A. Targeting the TGFβ signalling pathway in disease. Nat Rev Drug Discov. 2012;11:790-811.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1175]  [Cited by in F6Publishing: 1192]  [Article Influence: 91.7]  [Reference Citation Analysis (0)]
95.  Vancheri C, Sciacca E, Muscato G, Spicuzza L, Fruciano M, Gili E, Sambataro G, Palmucci S, Libra A. Pharmacological treatment in Idiopathic Pulmonary Fibrosis: current issues and future perspectives. Multidiscip Respir Med. 2024;19:982.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
96.  Ruiz-Ortega M, Lamas S, Ortiz A. Antifibrotic Agents for the Management of CKD: A Review. Am J Kidney Dis. 2022;80:251-263.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 37]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
97.  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. 2011;22:1144-1151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 210]  [Cited by in F6Publishing: 224]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
98.  Wu M, Chen W, Miao M, Jin Q, Zhang S, Bai M, Fan J, Zhang Y, Zhang A, Jia Z, Huang S. Anti-anemia drug FG4592 retards the AKI-to-CKD transition by improving vascular regeneration and antioxidative capability. Clin Sci (Lond). 2021;135:1707-1726.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
99.  Yu L, Border WA, Anderson I, McCourt M, Huang Y, Noble NA. Combining TGF-beta inhibition and angiotensin II blockade results in enhanced antifibrotic effect. Kidney Int. 2004;66:1774-1784.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 71]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
100.  Chen J, Zhang J, Shaik NF, Yi B, Wei X, Yang XF, Naik UP, Summer R, Yan G, Xu X, Sun J. The histone deacetylase inhibitor tubacin mitigates endothelial dysfunction by up-regulating the expression of endothelial nitric oxide synthase. J Biol Chem. 2019;294:19565-19576.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 19]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
101.  Zhao L, Han F, Wang J, Chen J. Current understanding of the administration of mesenchymal stem cells in acute kidney injury to chronic kidney disease transition: a review with a focus on preclinical models. Stem Cell Res Ther. 2019;10:385.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 15]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
102.  Liu Y, Su YY, Yang Q, Zhou T. Stem cells in the treatment of renal fibrosis: a review of preclinical and clinical studies of renal fibrosis pathogenesis. Stem Cell Res Ther. 2021;12:333.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 15]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
103.  Lang H, Dai C. Effects of Bone Marrow Mesenchymal Stem Cells on Plasminogen Activator Inhibitor-1 and Renal Fibrosis in Rats with Diabetic Nephropathy. Arch Med Res. 2016;47:71-77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
104.  Hsu YH, Chiu IJ, Lin YF, Chen YJ, Lee YH, Chiu HW. Lactoferrin Contributes a Renoprotective Effect in Acute Kidney Injury and Early Renal Fibrosis. Pharmaceutics. 2020;12:434.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
105.  Chen DQ, Chen L, Guo Y, Wu XQ, Zhao TT, Zhao HL, Zhang HJ, Yan MH, Zhang GQ, Li P. Poricoic acid A suppresses renal fibroblast activation and interstitial fibrosis in UUO rats via upregulating Sirt3 and promoting β-catenin K49 deacetylation. Acta Pharmacol Sin. 2023;44:1038-1050.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 15]  [Reference Citation Analysis (0)]
106.  Noel S, Kapoor R, Rabb H. New approaches to acute kidney injury. Clin Kidney J. 2024;17:65-81.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
107.  Parikh CR, Mansour SG. Perspective on Clinical Application of Biomarkers in AKI. J Am Soc Nephrol. 2017;28:1677-1685.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 54]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
108.  Murphy-Ullrich JE, Suto MJ. Thrombospondin-1 regulation of latent TGF-β activation: A therapeutic target for fibrotic disease. Matrix Biol. 2018;68-69:28-43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 175]  [Cited by in F6Publishing: 203]  [Article Influence: 29.0]  [Reference Citation Analysis (0)]