ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Sickle cell disease effects on the kidney

Sickle cell disease effects on the kidney
Literature review current through: Jan 2024.
This topic last updated: May 31, 2023.

INTRODUCTION — Sickle cell disease (SCD) refers to a group of related hemoglobinopathies in which the sickle hemoglobin mutation is co-inherited with another beta globin mutation (eg, Hb SS, sickle-beta thalassemia, Hb SC disease) leading to sickling and vaso-occlusion. (See "Overview of compound sickle cell syndromes".)

Kidney injury in SCD, referred to as sickle cell nephropathy (SCN), is a common, under-recognized complication. In addition, there are numerous other potential effects of SCD on the kidney and urinary tract.

This topic reviews the effects of SCD on the kidney and our approach to monitoring, prevention, and treatment. Separate topic reviews discuss the pathophysiology of vaso-occlusion, other clinical manifestations of SCD, and our overall approach to management.

Pathophysiology – (See "Pathophysiology of sickle cell disease".)

Clinical features – (See "Overview of the clinical manifestations of sickle cell disease".)

Management – (See "Sickle cell disease in infancy and childhood: Routine health care maintenance and anticipatory guidance" and "Sickle cell disease (SCD) in adolescents and young adults (AYA): Transition from pediatric to adult care" and "Overview of the management and prognosis of sickle cell disease" and "Sickle cell disease in sub-Saharan Africa".)

Effects of sickle cell trait on the kidney are also discussed separately. (See "Sickle cell trait", section on 'Urologic and kidney disease'.)

SICKLE CELL NEPHROPATHY

Pathogenesis

Features of SCN — SCD has several related effects on the kidney, collectively referred to as "sickle cell nephropathy" (SCN) or "sickle nephropathy"; kidney damage is multifactorial and often only becomes apparent retrospectively after the serum creatinine increases above the reference range [1-3].

The major site of kidney injury is the renal medulla, which is supplied by the vasa recta capillaries (figure 1). These capillaries create an environment that is especially conducive to sickling because it is relatively hypoxic as well as acidic and hypertonic [4].

Hypoxia, acidosis, and hypertonicity all increase sickling by promoting sickle hemoglobin polymerization via effects on hemoglobin structure; hypertonicity may also affect hemoglobin concentration within the cell.

Sickling in turn affects the vasculature in a number of ways, including increasing adhesion of red blood cells (RBCs) to the vascular wall; increasing inflammation; increasing vascular tone (ie, causing vasoconstriction, as free hemoglobin from hemolysis sequesters nitric oxide [NO]); and activating platelets and coagulation factors [2,5,6]. The role of simple obstruction of blood flow by sickled cells, previously thought to be the primary mechanism of vaso-occlusion, may contribute but does not occur in isolation. This subject is discussed in more detail separately. (See "Pathophysiology of sickle cell disease".)

These vascular effects can cause renal medullary ischemia and infarction, with gradual loss of glomerular and tubular function [7]. Sickling in the vasa recta is also thought to lead to the loss of deep juxtamedullary nephrons and reduced countercurrent exchange in the inner medulla, in turn impairing free water reabsorption and urinary concentrating ability; hyposthenuria is common [8-10]. Low urine osmolality in SCD did not consistently improve with vasopressin, which suggests factors beyond hyposthenuria may contribute to volume depletion, nocturia, and polyuria [11]. (See 'Enuresis' below.)

In contrast, the superficial loops of Henle in the cortical nephrons are supplied by peritubular capillaries, which do not experience the same degree of acidification, hypoxia, and hypertonicity.

The progression of changes and clinical features that appear over time are discussed below. (See 'Progression to clinically apparent findings' below.)

Biomarkers — Some of the observed abnormalities have led to their investigation as possible disease biomarkers [12]. However, none of these markers have been validated clinically and none are being used outside of clinical trials. Examples of markers under study include the following:

Proteins that might suggest hypoxia, hemolysis, and/or increased reactive oxygen species (eg, hypoxia-inducible factor 1 [HIF-1]; urinary and/or plasma endothelin 1 [ET-1]; serum lactate dehydrogenase [LDH] or asymmetric dimethylarginine [ADMA]), hemoglobinuria, or the kidney Doppler sonography resistive index and pulsatility index [13-19].

Mediators of inflammation or vascular changes (eg, urinary transforming growth factor [TGF]-beta 1; urinary kallikrein; quinolinic acid [QA]; soluble serum fms-like tyrosine kinase 1 [sFlt-1]; or the ratio of serum Fas to serum Fas ligand [sFas/sFasL]) [16,18,20-23].

Factors related to kidney injury or renal clearance (eg, serum cystatin C; urinary kidney injury molecule [KIM-1], N-acetyl-beta-D-glucosaminidase [NAG], beta2-microglobulin, BMPR1B, non-muscle myosin heavy chain 9 [MYH9], apolipoprotein 1 [APOL1], and others) [12,24-29]. (See "Gene test interpretation: APOL1 (chronic kidney disease gene)".)

Polymorphisms of the HMOX1 gene, which encodes the heme-oxygenase-1 enzyme, are also likely to affect the frequency of kidney disease [4].

Variants in APOL1 appear to only account for a small part of kidney disease morbidity, suggesting there may be important polymorphisms that have not yet been identified. A 2022 genome-wide association study (GWAS) identified several new loci, many with evidence for regulating proteinuria (CRYL1, VWF, ADAMTS7) and kidney disease (LRP1B, linc02288, and FPGT-TNNI3K/TNNI3K) [30]. The strongest overall association was with CRYL1.

Progression to clinically apparent findings — As with other manifestations of SCD, the severity and age of onset of SCN is variable (figure 2) [31]. SCN appears to be more severe with more severe genotypes (homozygous Hb SS and sickle-beta0 thalassemia) than with milder genotypes (Hb SC disease or sickle-beta+ thalassemia) [6,32]. However, there is significant variability even among individuals with the same genotype. (See 'Prevalence' below.)

The following progression of changes is typical:

Early changes – In infancy and early childhood, there may be increased glomerular pressure leading to compensatory kidney hypertrophy, glomerular hyperfiltration, and impaired urinary concentrating ability; creatinine is usually within the reference range [33-35]. Glomerular hyperfiltration is typically defined as an estimated glomerular filtration rate (eGFR) above a threshold value (eg, >130 mL/min per 1.73 m2 for women and >140 mL/min per 1.73 m2 for men) [5].

These changes may also be present in young adults; in one study, approximately one-half of adults <40 years of age had evidence of hyperfiltration [5]. Glomerular hyperfiltration is usually transient, followed by a decline in GFR over the ensuing years, as illustrated in a case series and an animal model [36,37]. It is not known whether hyperfiltration predicts the later development of chronic kidney disease (CKD) [5,38]. (See "Secondary factors and progression of chronic kidney disease", section on 'Intraglomerular hypertension and glomerular hypertrophy'.)

Later childhood – Later in childhood or early adulthood, microalbuminuria, hematuria, or both may develop. The predominant left-sided origin of the hematuria has been attributed to the greater length of the left renal vein and the "nutcracker phenomenon," in which compression of the left renal vein between the aorta and superior mesenteric artery leads to increased blood pressure in the vein [39].

Hyperfiltration – Hyperfiltration, defined as eGFR >180 mL/min/1.73m2, is common and occurs early in children with SCD [40]. Hyperfiltration typically precedes albuminuria [36]. In one study, baseline GFR measured quantitatively by technetium 99m-labeled diethylenetriaminepentaacetic acid (DTPA) plasma clearance was performed in 184 infants (mean age, 13 months) [41]. The average DTPA GFR was 125.2 mL/min/1.73m2 (compared to a normal value of 91.5 mL/min/1.73m2). DTPA GFR was correlated with age, weight, height, and kidney volume, but not with hemoglobin. Data from a second study in children with SCD indicated that hydroxyurea at maximum tolerated dose is associated with a decrease in hyperfiltration in young children with SCD [40].

As expected, because hyperfiltration occurs in children <1 year of age, there is a high rate hyperfiltration among adults. In a series of 280 adults with SCD, the prevalence of hyperfiltration assessed by Modification of Diet in Renal Disease (MDRD)-estimated GFR was 51 percent [5]. Among adults with hyperfiltration, approximately one-half had hyperfiltration alone, whereas smaller numbers had associated microalbuminuria or macroalbuminuria (36 and 15 percent, respectively). In a subgroup of 48 patients who underwent GFR measurement using a radioactive tracer, the sensitivity of the estimated GFR for hyperfiltration was 94 percent, and the specificity was 63 percent.

Proximal tubular function – Proximal tubular function is also supranormal [42]. This may be manifested by hyperphosphatemia (due to increased phosphate reabsorption) and elevated creatinine clearance (due to enhanced creatinine secretion) that does not reflect the true GFR. The increased reabsorption of sodium and other solutes results in high levels of phosphorus, beta-2-microglobulin, and uric acid [34,43-45]. (See 'Accurate estimation of kidney function' below.)

Distal tubular function – Also by an unclear mechanism, there may be impaired distal hydrogen and potassium secretion, leading to a partial distal renal tubular acidosis [34,43,46-48]. Reduced potassium and hydrogen ion excretion can result in inadequate urinary acidification and cause metabolic acidosis in settings of acute illness or excess acid load [45]. The distal renal tubular acidosis is progressive, increases with age, and correlates with a decreasing GFR, plasma bicarbonate, urinary osmolarity, and hyperkalemia.

Proposed explanations include medullary blood flow disturbance and hypoxia leading to reduced hydrogen ion and electrochemical gradients along the collecting ducts. Consequences may include hyperkalemia, which is typically mild and only becomes clinically apparent if there is a complicating factor such as potassium or acid loading, intravascular volume depletion, or rhabdomyolysis. (See "Overview and pathophysiology of renal tubular acidosis and the effect on potassium balance".)

Kidney biopsy generally is not performed, but reports of pathology in the early stages have described glomerular enlargement, hemosiderin deposits, papillary necrosis, and cortical infarction [8,35].

Early adulthood – In early adulthood, macroalbuminuria and reduced GFR may be seen; however, creatinine may remain within the reference range because of the increased creatinine clearance.

Microalbuminuria is an early clinical manifestation of glomerular damage. It increases with age and affects 60 percent of SCD patients >45 years of age [11]. In addition to age, albuminuria is associated with anemia and hemolysis.

Albuminuria is a risk factor for accelerated decline in kidney function.

In later adulthood, there is progressive reduction in GFR and in some cases development of end-stage kidney disease (ESKD), as glomerular hypertrophy can no longer compensate for vascular dropout (image 1). At this point, the serum creatinine will be increased.

Less commonly, membranoproliferative glomerulonephritis (MPGN) with mesangial expansion and basement membrane duplication may be seen, either as an isolated finding or in association with focal segmental glomerulosclerosis (FSGS) [49,50]. It has been proposed that this form of MPGN is caused by fragmented RBCs lodged in isolated capillary loops and phagocytosed by mesangial cells, stimulating expansion of the mesangium and new basement membrane deposition [8,51]. In contrast with immune complex- and complement-mediated MPGN, in SCD there are no immune complexes or electron-dense deposits.

Reports of pathology in later stages have described interstitial inflammation, edema, fibrosis, tubular atrophy, papillary infarcts, and progressive glomerulosclerosis [8,35,52]. Unlike analgesic nephropathy, in which vasa recta are often spared, in SCN there is destruction of the vasa recta and vascular congestion of the collecting ducts, inner medulla, and papillae [53]. There is progressive nephron damage and FSGS (perihilar [most common] or collapsing), and the vasa recta are almost completely lost in some older individuals [35,49,52-54]. (See "Focal segmental glomerulosclerosis: Pathogenesis".)

Rapid clinical deterioration – In a longitudinal study of 169 consecutive adults with SCD followed for over 13.5 years, there was a strong association between rapid decline in kidney function and SCD disease severity, especially with the SCD "hemolysis phenotype" [55]. This hemolytic phenotype associated with kidney disease progression has also been associated with pulmonary hypertension, systemic hypertension, and stroke [2]. Genetic factors that attenuate this hemolysis phenotype, such as co-inheritance of alpha thalassemia or the sickle cell-beta0 haplotype, will likely lower the risk of CKD. During long-term follow-up, plasma NT-proBNP, a known risk factor for mortality, was also significantly associated with declining kidney function.

Other morbidities contributing to declining kidney function — Other factors may contribute to kidney injury at any stage, as discussed in more detail separately. (See "Secondary factors and progression of chronic kidney disease".)

The following may be especially relevant to individuals with SCD:

Nephrotoxic medications – Individuals with SCD are likely to receive a number of potentially nephrotoxic agents. (See 'Avoiding medications that are toxic to the kidney' below.)

Dosing of these drugs may be inappropriately high in some instances such as when estimation of the GFR used to calculate drug dosing is inaccurate (see 'Accurate estimation of kidney function' below) or when high doses of nonsteroidal antiinflammatory drugs (NSAIDs) are used in an attempt to minimize opioid pain medications. (See "Clinical manifestations and diagnosis of analgesic nephropathy".)

Hypertension – Endothelial dysfunction can also lead to hypertension, which independently increases the risk of kidney disease. (See "Antihypertensive therapy and progression of nondiabetic chronic kidney disease in adults", section on 'Effect of goal blood pressure on progression of CKD'.)

Features that correlate with the risk of progressive kidney injury include hemoglobin genotype, age, hypertension, albuminuria, and low fetal hemoglobin (Hb F) [2-4,56,57]. In addition, variants in the APOL1 gene, which encodes apolipoprotein L1, are emerging as risk factors for kidney disease in individuals with SCD as well as African Americans without SCD [26,58-60]. (See 'Progression to clinically apparent findings' above.)

Prevalence — SCN leading to CKD occurs in as many as one-fourth to one-third of adults with SCD. In one study, 30 percent of adults with SCD developed CKD by age 31, and within five years, 42 percent had progressed to ESKD [61].

The prevalence of CKD is higher in individuals with Hb SS and sickle-beta0 thalassemia, but individuals with Hb SC disease also develop SCN at a relatively high rate.

A 2016 analysis determined the rates of kidney disease in adults with SCD enrolled in one of several clinical studies (Treatment of Pulmonary Hypertension and SCD with Sildenafil Therapy [Walk-PHaSST], Cooperative Study of SCD [CSSCD], and Multicenter Study of Hydroxyurea in SCD [MSH]) [32]. Collectively, these trials included 1470 individuals with homozygous Hb SS. The Walk-PHaSST trial was the most recent of these trials, conducted from 2007 to 2009, and it also included 127 individuals with Hb SC disease. Key findings included the following:

Age was inversely correlated with eGFR (average loss of approximately 1.7 to 1.8 mL/min/1.73 m2 per year in Hb SS; approximately 1.1 mL/min/1.73 m2 per year in Hb SC). By comparison, age-related decline in eGFR in individuals without SCD is approximately 0.5 mL/min/1.73 m2 per year.

Moderately increased albuminuria (formerly called "microalbuminuria") was seen in 44 percent of those with Hb SS and 23 percent of those with Hb SC; severely increased albuminuria (formerly called "macroalbuminuria") was seen in 20 percent (both genotypes).

A 2016 Medicaid database review of 9481 individuals with SCD hospitalized during the period from 2007 to 2012 documented the presence of CKD in 0.1 percent of children, 5 percent of adults, and 15.9 percent of adults over the age 40 years [62]. CKD was seen in 68 percent of admissions with hypertension and 25 percent of admissions with heart failure. Over the course of the study, new CKD was seen in 6.7 percent (annual incidence, 1.3 percent), which was approximately two- to threefold higher than in a non-SCD control group. Various smaller series have reported estimates of CKD in the range of 21 to 29 percent of adults with SCD [56,57].

A 25-year longitudinal study from 1991 involving 725 individuals with Hb SS and 209 with Hb SC reported CKD in 4.2 and 2.4 percent, respectively [53]. The median age at which CKD occurred was 23 years for Hb SS and 50 years for Hb SC disease.

A 2022 retrospective study of 2,194,079 patients enrolled in the United States Renal Data System (USRDS) registry found that ESKD was diagnosed at a younger age in individuals with SCD compared with individuals without SCD, with 20 percent of patients with SCD developing ESKD before age 30 [61]. Patients with SCD had a higher mortality (2.7-fold higher) and a lower transplant rate than those without SCD.

In a cohort of 870 patients, older age, male sex, higher diastolic blood pressure, and anemia were risk factors for a GFR <90 mL/min/1.73 m2, which appeared to show a trend towards increased mortality, especially in those with GFR <60 mL/min/1.73 m2, which correlated with a 12-fold increase in mortality [63].

Evaluation

Routine surveillance and early detection — The development of CKD can be insidious, and ongoing surveillance is standard of care and an essential component of management for all individuals with SCD [2,3]. This is part of a larger program of care that includes a number of other routine assessments and preventive interventions. (See "Overview of the management and prognosis of sickle cell disease", section on 'Routine evaluations and treatments' and "Sickle cell disease in infancy and childhood: Routine health care maintenance and anticipatory guidance".)

While surveillance is essential, high-quality evidence is lacking to guide the optimal tests and intervals for testing. Typically, routine evaluations are initiated by age three to five years and no later than 10 years of age. Routine visits occur approximately two to three times per year for children and approximately four to six times per year for adults. We typically do the following at every visit:

Assessment of blood pressure. (See 'Blood pressure abnormalities' below.)

At least annually, we check a urinalysis with examination of the urinary sediment and a spot urine albumin-to-creatinine ratio [64].

If the urine albumin-to-creatinine ratio is above 300 mg/g, a 24-hour urine collection for protein quantification should be performed; typically this is done by the consulting nephrologist. We often repeat the urine albumin-to-creatinine ratio before referral to nephrology in order to limit unnecessary referrals. Treatment of proteinuria is discussed below. (See 'Nephrologist referral and CKD interventions' below.)

If microscopic hematuria is present, it is evaluated as discussed below. (See 'Evaluation of hematuria and flank pain' below.)

A metabolic panel including serum creatinine, which is used to calculate the eGFR. Some experts recommend using the Schwartz formula, and others use other formulas such as the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) (calculator 1) to calculate eGFR. (See 'Accurate estimation of kidney function' below.)

Hydroxyurea can interfere with serum creatinine measurement by the i-STAT device; patients receiving hydroxyurea who have an increased serum creatinine measurement using the i-STAT device should have a repeat measurement by another method. (See 'Role of hydroxyurea and transfusions' below.)

Individuals with SCD can develop proximal and distal renal tubular acidosis (RTA) that is characterized by a metabolic acidosis with a low carbon dioxide (CO2) and abnormal potassium levels, and occasionally hyperchloremia [65]. Iron chelating agents may cause Fanconi syndrome (generalized proximal tubular dysfunction), which may affect calcium and phosphate levels in the blood and urine [66,67]. These individuals should have correction of their acidosis with bicarbonate as well as appropriate treatment for the RTA. (See "Etiology and clinical manifestations of renal tubular acidosis in infants and children", section on 'Fanconi syndrome'.)

If the eGFR declines by more than 10 percent in a given year or drops below 60 mL/min/1.73 m2, we evaluate the urine sediment and perform kidney imaging [3].

Patients with SCD might already have substantial kidney impairment at the time of diagnosis using creatinine and/or GFR, which underestimates the degree of kidney injury. Cystatin C is an alternative biomarker and not influenced by muscle mass or creatinine diet and is not secreted by the proximal tubules. It appears to be useful in reflecting the causal effects of hemolysis and associated deficiency of hemolysis scavenger proteins in patients with kidney dysfunction. A Jamaican study found that cystatin C correlated better than the serum creatinine with kidney dysfunction [11]. While it appears useful, it has not been validated for routine testing and is not universally available, and more studies are needed before it is routinely recommended.

In research settings, technetium-DTPA measurements have been used; however, DTPA may underestimate GFR due to plasma protein binding.

Kidney imaging (eg, ultrasound) is appropriate and part of the routine evaluation for individuals with SCD who have an acute or chronic decline in kidney function (eg, by more than 10 percent), significant hematuria or proteinuria, or any clinical symptoms or other findings consistent with urinary obstruction. The main purpose is to identify obstructive uropathy or other anatomic abnormalities. As noted below, ultrasound is not sensitive for renal medullary carcinoma, and individuals with persistent hematuria or other concerning symptoms should have computed tomography (CT) imaging. (See 'Evaluation of hematuria and flank pain' below.)

Certain individuals require more frequent monitoring of kidney function (eg, individuals receiving iron chelation with deferasirox should have a monthly metabolic panel and urine albumin and creatinine; those receiving deferoxamine should have this testing every three months). Dose reduction or temporary discontinuation may be required for significant increases in creatinine. (See 'Avoiding medications that are toxic to the kidney' below and "Transfusion in sickle cell disease: Management of complications including iron overload", section on 'Chelation therapy'.)

This approach is largely consistent with guidance from a National Heart, Lung, and Blood Institute (NHLBI) expert panel, which recommends initial screening for proteinuria beginning by age 10 years. We start assessing for proteinuria at age five or six years because our clinical experience along with that of others demonstrates that persistent proteinuria can occur before the age of 10 years [36]. We suggest annual surveillance for those without evidence of proteinuria and referral of individuals with proteinuria (urine albumin-to-creatinine ratio of >300 mg/g or >300 mg/24 hours) to a nephrologist [10,68]. The panel expressed some reservations regarding the appropriate testing but also presented a consensus view that many of the assessments for kidney disease have relatively low costs and burdens, and the potential for benefit is high if renal dysfunction is detected and treated early.

The usefulness of kidney biopsy has not been evaluated in SCD, and the decision to biopsy the kidney is individualized, similar to the general population [3,10]. As noted above, markers of early kidney disease are sought but have not been validated and are not used routinely. (See 'Pathogenesis' above and "The kidney biopsy", section on 'Indications'.)

Additional testing for atypical presentations — Individuals with atypical presentations (eg, more severe proteinuria, proteinuria plus hematuria, decline in hemoglobin out of proportion to severity of reduced kidney function) may require additional testing [3].

Examples include:

For individuals over age 40 who develop new or worsening kidney dysfunction, testing for multiple myeloma with a serum protein electrophoresis (SPEP) and urine protein electrophoresis, immunofixation, and serum free light chain assay is appropriate. (See "Laboratory methods for analyzing monoclonal proteins".)

For individuals with new-onset nephrotic syndrome, recent transient red cell aplasia, or liver function test abnormalities, testing for parvovirus B19, viral hepatitis (hepatitis B or C), and/or HIV may be helpful. (See "Glomerular disease: Evaluation and differential diagnosis in adults", section on 'Evaluation of nephrotic syndrome'.)

For individuals with systemic lupus erythematosus (SLE) or SLE-like features, testing for kidney involvement is done periodically, including urinalysis and examination of the urine sediment, anti-DNA antibody titers, and other markers of disease activity. Kidney biopsy is often performed. Details are presented separately. (See "Lupus nephritis: Diagnosis and classification", section on 'Evaluation and diagnosis'.)

Diagnosis — The diagnosis of SCN is made in an individual with kidney dysfunction (decline in eGFR, proteinuria) without another explanation after a thorough evaluation for other causes of these findings. It is a diagnosis of exclusion. The extent of the evaluation depends on the patient's age and clinical status as discussed above. (See 'Routine surveillance and early detection' above and 'Additional testing for atypical presentations' above.)

Accurate estimation of kidney function — Accurate determination of kidney function is critical to determining the need for interventions. Calculation of the eGFR as discussed above is used to estimate kidney function in patients with SCD. (See 'Routine surveillance and early detection' above.)

Potential sources of interference in estimating GFR in SCD include the following:

Glomerular hyperfiltration and supranormal proximal tubule function may lower the serum creatinine and suggest kidney function is better than it is. In a study involving 19 individuals with SCD, the creatinine clearance overestimated the measured GFR by 29 percent (154 versus 119 mL/min), roughly twice the percent difference in a group of eight age- and sex-matched controls [69]. The Schwartz formula was designed to minimize such inaccuracy in estimating GFR; however, it has limitations. Even the CKD-EPI equation was shown to have overestimated GFR by approximately 45 mL/min/1.73 m2 [70]. (See 'Pathogenesis' above.)

Hydroxyurea interferes with one device (the i-STAT device) that measures serum creatinine and is used in certain clinical laboratories. (See 'Role of hydroxyurea and transfusions' below.)

Other medications that may falsely affect the estimated GFR are listed separately. (See "Drugs that elevate the serum creatinine concentration".)

Cystatin C may be useful in some individuals and may correlate better with GFR, although it has not been validated specifically in SCD across different age groups. In a series of 98 individuals with SCD, cystatin C was well-correlated with measured GFR [71]. The cystatin C-based CKD-EPI showed the greatest agreement with GFR compared with other CKD-EPI equations.

This subject is discussed separately. (See "Chronic kidney disease in children: Clinical manifestations and evaluation", section on 'Serum creatinine and glomerular filtration rate' and "Assessment of kidney function" and "Calculation of the creatinine clearance".)

Prevention and management (sickle nephropathy) — Lack of access to early multidisciplinary care contributes to increased mortality in SCD, and a significant proportion of this early mortality is related to sickle nephropathy. It is especially important to ensure that individuals with SCD have timely access to preventive therapies and to replacement therapy (dialysis or kidney transplantation) when needed.

Avoiding medications that are toxic to the kidney — The following medications are often used in SCD and are potentially nephrotoxic:

NSAIDs – Nonsteroidal antiinflammatory drugs (NSAIDs), including ketorolac, may be over-used for pain. In some cases, short-term use of NSAIDs may be appropriate; however, routine use of NSAIDs for fever, minor pain, or as an adjunct to opioids, should generally be avoided due to the increased risks of acute and chronic kidney injury. Families and caregivers should be counseled to use acetaminophen instead of NSAIDs for fever and routine minor pains.

Vaso-occlusive pain management is an especially challenging area for individuals with SCD. Opioid pain medications and other means of pain control should be available and given in appropriate doses as needed. High doses of NSAIDs, especially ketorolac, should be minimized or avoided during hospitalization. Hydroxyurea may reduce the frequency of painful episodes but can take months to become effective, and appropriate pain medications should not be withheld while awaiting the beneficial effects of hydroxyurea. (See "Acute vaso-occlusive pain management in sickle cell disease", section on 'Therapies we do not use'.)

Aminoglycoside antibiotics – Broad spectrum antibiotics including vancomycin and gentamicin are often required for fever, which could be a sign of life-threatening infection. Gram-positive coverage may be especially important in those with indwelling venous catheters. The serum creatinine is a useful test for estimating eGFR, but caveats should be kept in mind. (See 'Accurate estimation of kidney function' above and "Evaluation and management of fever in children and adults with sickle cell disease", section on 'Empiric antibiotic therapy'.)

Close attention to dosing and drug levels is essential to prevent drug accumulation. Attention to volume status is also important. (See "Pathogenesis and prevention of aminoglycoside nephrotoxicity and ototoxicity".)

Radiocontrast material – Contrast-enhanced imaging studies may be needed to evaluate suspected acute chest syndrome, splenic or hepatic sequestration, or stroke. Radiocontrast material used in CT scans can also increase the risk for acute kidney injury (AKI) in patients with severely impaired kidney function (eGFR <30 mL/min/1.73 m2); close attention should be paid to adequate hydration and using the smallest dose necessary. (See "Contrast-associated and contrast-induced acute kidney injury: Clinical features, diagnosis, and management" and "Patient evaluation prior to oral or iodinated intravenous contrast for computed tomography", section on 'Patients with impaired kidney function'.)

Iron chelators – Chelation therapy to remove excess iron stores (typically from transfusional iron overload) are often needed. The oral iron chelator deferasirox causes a known dose-dependent rise in serum creatinine, but this appears to be reversible with close monitoring and discontinuation if the creatinine rises more than 50 percent over baseline or more than two times the upper limit of the reference range. Dose reduction is used for smaller increases in creatinine. Deferasirox also has a Boxed Warning stating that it can cause AKI, especially in individuals with comorbidities. Details are discussed separately. (See "Transfusion in sickle cell disease: Management of complications including iron overload", section on 'Chelation therapy'.)

Others – Other medications to be avoided, if possible, include certain chemotherapeutic agents, amphotericin, beta-lactam antibiotics, and certain immunosuppressive drugs [72].

The anticoagulant edoxaban (a direct factor Xa inhibitor) is not nephrotoxic, but there are concerns with its use in individuals with hyperfiltration (creatinine clearance >95 mL/min). If anticoagulation is needed for venous thromboembolism (VTE) or atrial fibrillation, we make sure to adhere to specific dosing recommendations or to use a different anticoagulant. (See "Direct oral anticoagulants (DOACs) and parenteral direct-acting anticoagulants: Dosing and adverse effects", section on 'Edoxaban'.)

Role of hydroxyurea and transfusions — Hydroxyurea is a mainstay of SCD therapy and is recommended for infants as young as nine months of age who have homozygous hemoglobin SS or sickle-beta0 thalassemia. High-quality evidence has demonstrated reduction of several types of vaso-occlusive events. Further details of its mechanism of action, evidence for efficacy, administration, and adverse effects are presented separately. (See "Hydroxyurea use in sickle cell disease".)

For those not receiving hydroxyurea, it is important that they be made aware of the potential benefits as well as the risks; in some cases, the perceived risks may not be supported by available evidence. (See "Hydroxyurea use in sickle cell disease", section on 'Adverse effects'.)

Evidence for improvement in kidney function with hydroxyurea therapy comes from observational studies and indirect outcomes in randomized trials. As examples:

Infants and children – The BABY HUG trial, which involved 193 infants (median age, 13.8 months) with sickle cell anemia treated with hydroxyurea or placebo for two years, found indirect evidence of improved kidney function with hydroxyurea (increased urine osmolality and decreased kidney volume on ultrasound) but no significant effect on GFR [73]. Another study suggested that hydroxyurea reduces hyperfiltration in children [40].

Adults – In a series of 112 adults in a comprehensive sickle cell program, those receiving hydroxyurea had lower rates of albuminuria (35 percent versus 55 percent in those not receiving hydroxyurea) [74].

Other studies and case reports have also described improvement in kidney function with hydroxyurea and worsening of kidney function when hydroxyurea was discontinued [3,75].

Hydroxyurea can be coadministered with an angiotensin-converting enzyme (ACE) inhibitor or an angiotensin II receptor blocker (ARB); in fact, the combination of hydroxyurea plus one of these agents may lead to further reduction of proteinuria [74,76,77].

Hydroxyurea is metabolized by the kidney and removed by hemodialysis; dose adjustments may be needed in individuals with reduced kidney function. It has been recommended that individuals undergoing hemodialysis who are initiating hydroxyurea use a lower starting dose (eg, 7.5 mg/kg daily rather than 15 mg/kg daily) [7]. On dialysis days, hydroxyurea should be given after dialysis. Information about dose titration is presented separately. (See "Hydroxyurea use in sickle cell disease", section on 'Monitoring and dose titration'.)

Clinicians should be aware that patients receiving hydroxyurea may have falsely elevated creatinine measurement when using a point-of-care device for measuring serum creatinine (the i-STAT system, used for routine testing in some clinical laboratories) [78]. In such cases, a method of creatinine measurement other than the i-STAT should be used. (See 'Accurate estimation of kidney function' above.)

Transfusions may be beneficial in preventing kidney injury. In the TWiTCH trial, which compared transfusion to no transfusion, patients assigned to transfusion did not have progression of kidney disease and had less albuminuria [79].

Transfusions are not used to treat AKI or CKD. However, case reports have described improvement or stabilization of renal function in selected individuals treated with regular transfusions for other indications [3,80]. In some cases, individuals with SCD and ESKD who have severe anemia that is unresponsive to erythropoiesis-stimulating agents (ESAs) may be treated with transfusions while awaiting other therapies [3]. (See 'ESKD interventions (dialysis and transplant)' below.)

Nephrologist referral and CKD interventions — Referral to a nephrologist may be appropriate for any of the following [10]:

Decline in kidney function or development of proteinuria (eg, positive urine dipstick for protein), even if eGFR remains within the reference range

eGFR below 60 mL/min/1.73 m2

Any degree of albuminuria in a child

Hematuria, especially if persistent

Hypertension or relative hypertension

There have been very few randomized trials addressing optimal therapy in individuals with proteinuria and hypertension [81]. However, available evidence supports the use of an ACE inhibitor or an ARB:

A small trial randomly assigned 22 individuals with SCD who had moderately increased albuminuria and normal blood pressure to receive captopril 25 mg daily or placebo for six months [82]. The captopril group had a small decrease in proteinuria (by 45 mg per 24 hours), while the placebo group had an increase in proteinuria (by 18 mg per 24 hours). The captopril group had a small change in blood pressure.

Several observational studies have also described improvements in moderately increased albuminuria, less rapid decline in eGFR in individuals with SCD when treated with ACE inhibitors or ARBs, and worsening when ACE inhibitors were discontinued [52,83,84]. One of these was a retrospective study of 86 individuals with SCD who were followed for over two years while receiving one of these agents compared with 68 individuals not receiving an agent to block the renin-angiotensin-aldosterone system; the study demonstrated a reduction in the development in CKD and a trend towards reduced proteinuria associated with the ACE inhibitor or ARB [84].

There is good evidence in the non-SCD population that reductions in moderately increased albuminuria (for individuals with >500 to 1000 mg/day proteinuria) and control of blood pressure with an ACE inhibitor or an ARB slows the progression to ESKD, and the same benefits would be expected in SCD. (See "Antihypertensive therapy and progression of nondiabetic chronic kidney disease in adults", section on 'Effect of antihypertensive drugs on proteinuria'.)

Kidney failure is a morbid, common complication in SCD. Evidence-based, long-term trials of the benefit of treating proteinuria in SCD are not available. However, our consensus recommendations are to initiate treatment with an ACE inhibitor or ARB when there is persistent urine protein >500 mg per day, and especially when the urine protein to creatinine ratio (uPCR) is >100 mg/mmol (>884 mg/g), which is approximately equivalent to urine protein ≥1000 mg per day. This treatment includes patients who do not have hypertension. (See "Antihypertensive therapy and progression of nondiabetic chronic kidney disease in adults".)

Close monitoring of the blood pressure is essential to avoid hypotension. Supporting evidence in the non-SCD population, goals of therapy, details of administration, and adverse effects are presented separately; it is especially important to monitor the blood pressure closely in individuals with SCD because these individuals often have low blood pressure. (See "Antihypertensive therapy and progression of nondiabetic chronic kidney disease in adults" and "Goal blood pressure in adults with hypertension", section on 'Patients with chronic kidney disease' and 'Hypotension' below.)

Other interventions to correct metabolic abnormalities (hydration, calcium supplementation, phosphate binders, potassium restriction) and to slow the rate of CKD progression (protein restriction, smoking cessation, and glycemic control), as well as treatment of the complications of kidney failure (volume overload, neuropathy, uremic bleeding, pericarditis) are similar to individuals without SCD and are discussed in detail separately. (See "Chronic kidney disease in children: Overview of management" and "Overview of the management of chronic kidney disease in adults".)

Regular transfusions have been suggested for individuals with worsening kidney function approaching the need for kidney transplantation, especially if their anemia is no longer responsive to ESAs [3]. In a cohort study of African American ESKD patients initiating dialysis during the period from 2000 to 2014, high-dose ESA and severity of anemia were both risk factors for worse outcomes for ESKD [85].

While transfusions are avoided in the non-SCD population to minimize the risk of immunologic sensitization and potential adverse effects on future transplantation, this is not realistic in most people with SCD, who are often transfused for a number of complications. Transfusions should be used when indicated, with appropriate precautions to minimize alloimmunization; these are discussed in more detail separately. Leukocyte-reduced blood products have been demonstrated to reduce the risk of antibody-mediated kidney allograft rejection [86]. (See "Red blood cell transfusion in sickle cell disease: Indications and transfusion techniques", section on 'Indications for transfusion' and "Red blood cell transfusion in sickle cell disease: Indications and transfusion techniques", section on 'Transfusion techniques'.)

ESKD interventions (dialysis and transplant) — Replacement therapy (dialysis or kidney transplantation) is appropriate for individuals with SCD who develop ESKD, similar to the general population. (See "Indications for initiation of dialysis in chronic kidney disease" and "Kidney transplantation in adults: Evaluation of the potential kidney transplant recipient".)

Dialysis – Individuals with SCD and ESKD can be treated successfully with hemodialysis or peritoneal dialysis [3]. Dialysis is generally initiated at a younger age in individuals with SCD than in the general population. In a cohort of 397 individuals with SCN, the median age at which ESKD developed was 41±14 years [87]. The choice between hemodialysis and peritoneal dialysis depends on clinical factors and patient preferences. For those who are also receiving transfusions, the same vascular access can be used for hemodialysis; discussion between the nephrologist and hematologist before placement of dialysis access or a venous catheter for transfusion is advised to ensure that the device used is appropriate for both. The incidence of hemodialysis-related complications does not appear to be different in SCD compared with the general population, although survival appears to be decreased [7,88].

SCD and hemodialysis are both risk factors for pulmonary hypertension. This high risk should be addressed by evaluation of pulmonary status before placement of dialysis access. Separate topic reviews discuss the symptoms of pulmonary hypertension (eg, dyspnea, chest pain, exercise intolerance) and the appropriate evaluations. (See "Pulmonary hypertension associated with sickle cell disease" and "Pulmonary hypertension in patients with end-stage kidney disease" and "Overview of the management and prognosis of sickle cell disease", section on 'Routine evaluations and treatments'.)

Kidney transplantation – Kidney transplantation offers the best outcome for individuals with ESKD from SCN and should be offered to all potential candidates [89,90].

In a 2009 series involving 237 individuals with SCD who underwent kidney transplantation, the 10-year survival was 56 percent [91]. By comparison, those with ESKD who did not receive a transplant had a 10-year survival of 14 percent. In a subsequent study that compared survival rates over time, the six-year survival increased between an earlier era (from 1988 to 1999; median survival, 56 percent) to a later era (from 2000 to 2011; median survival, 70 percent) [92].

A large case series of individuals with ESKD reported that those with SCD were less likely to be placed on a kidney transplant waiting list, which may account for decreased survival from ESKD in the SCD population [87].

Several explanations exist for the lower rates of transplantation in SCD, including racial bias, concerns about higher infectious risk, concerns about blood group incompatibility that may lead to rejection, concerns about pulmonary hypertension, and concerns about side effects of immunosuppressive drugs (eg, avascular necrosis of bone as a complication of glucocorticoids); none of these should be reasons for avoiding transplantation [93]. (See 'Prognosis' below.)

Pulmonary hypertension in kidney transplant candidates is discussed separately. (See "Kidney transplantation in adults: Evaluation of the potential kidney transplant recipient", section on 'Pulmonary disease'.)

Recommended perioperative interventions include using transfusion to a preoperative hemoglobin level of no more than 10 g/dL; using exchange transfusion to avoid hyperviscosity when appropriate; minimizing alloimmunization by using antigen-matched blood; and paying close attention to oxygenation, hydration, respiratory status, and pain control [94]. Attention must be paid to the increased risks of VTE and the greater risk of infection related to functional asplenia. ESKD typically reduces the efficacy of vaccinations, and boosters may be needed prior to transplant [7]. Transfusion and infectious disease considerations are discussed in more detail separately. (See "Red blood cell transfusion in sickle cell disease: Indications and transfusion techniques" and "Overview of the management and prognosis of sickle cell disease", section on 'Infection prevention'.)

Typical graft survival at one year appears to be approximately 67 to 82 percent [94]. SCN can occur in the transplanted kidney unless the underlying disease process is altered (eg, with hematopoietic stem cell transplantation or disease-modifying therapies [91,95,96]).

As noted below and in separate topic reviews, evaluation for hyperkalemia and its appropriate treatment should be undertaken. (See 'Hyperuricemia and hyperkalemia' below and "Management of hyperkalemia in children" and "Treatment and prevention of hyperkalemia in adults".)

Anemia interventions — SCD causes chronic hemolytic anemia at baseline and lower erythropoietin and baseline hemoglobin levels [97]. The threshold for interventions to increase the hemoglobin level differs from non-SCD individuals. The optimal hemoglobin level for an individual with SCD and ESKD is unknown, and there are no good outcomes data for the use of ESAs.

For individuals with SCD and CKD who develop worsening anemia, especially with a decrease in reticulocyte count, use of an ESA such as epoetin or darbepoetin is appropriate. These medications can be given concurrently with hydroxyurea, and there is a theoretical possibility that concurrent use may reduce the risk of adverse events related to the ESA. Some studies have suggested that epoetin may trigger vaso-occlusion and/or VTE, although this was not borne out in a retrospective analysis [98].

ESA doses and schedules of administration have varied in different case series, as summarized in a review article from 2016 [7]. Choice of ESA, dosing, and adverse effects are presented in detail separately. (See "Treatment of anemia in nondialysis chronic kidney disease".)

Important caveats related to the use of ESAs in individuals with SCD include the following [7]:

Avoid hyperviscosity – Complications of hyperviscosity (vaso-occlusion, stroke, or VTE) may occur if the hemoglobin level is too high, especially in individuals with Hb SC disease. We generally try to avoid an increase in hemoglobin of more than 1 g/dL in a two-week period and generally target a hemoglobin level not higher than 10 g/dL (hematocrit 30 percent) when the Hb S is >35 percent of total hemoglobin. Some experts prefer to target a hemoglobin level that does not exceed 8 to 9 g/dL in individuals with frequent pain episodes or other frequent vaso-occlusive complications.

Avoid iron overload – Individuals with SCD often have iron overload and may be receiving iron chelation therapy. Routine treatment with supplemental iron is not appropriate; however, iron replacement therapy should be administered if an individual with SCD develops iron deficiency. (See "Treatment of iron deficiency in patients with nondialysis chronic kidney disease (CKD)" and "Treatment of iron deficiency in patients on dialysis" and "Transfusion in sickle cell disease: Management of complications including iron overload", section on 'Chelation therapy'.)

Individuals with SCD are also frequently treated with transfusions preoperatively or in the setting of certain acute vaso-occlusive complications. (See "Red blood cell transfusion in sickle cell disease: Indications and transfusion techniques".)

Investigational approaches — Preliminary studies suggest that drugs that decrease hemolysis, such as voxelotor, may safely reduce albuminuria [99,100].

Prognosis — SCN has been estimated to contribute substantially to morbidity and mortality in SCD.

ESKD accounts for approximately 16 to 18 percent of deaths in some series [6]. In a study from 1991, CKD developed in approximately 4 percent of individuals with Hb SS disease, and death occurred at approximately four years after the development of kidney failure (at a median age of 27 years) [53]. Median survival among patients with and without kidney failure was 29 and 51 years, respectively. The mortality risk of patients with kidney failure was similar to those who had had a stroke.

Survival is substantially decreased among patients with kidney failure and SCD compared with those with kidney failure without SCD. The unadjusted five-year risk of death in patients with SCD and ESKD was significantly elevated compared with the reference population (hazard ratio [HR], 2.00, 95% CI: 1.66-2.40) [85]. However, one analysis found that this reduced survival did not persist when adjusted for the lower rate of kidney transplantation in SCD [87,93]. Lack of access to multidisciplinary care may also contribute to worse survival [2]. Typical survival one year after kidney transplantation is approximately 88 percent [94].

These observations highlight the importance of comprehensive care during all stages of life as well as the need for disease-modifying therapies that can prevent complications of SCD. (See "Sickle cell disease in infancy and childhood: Routine health care maintenance and anticipatory guidance" and "Sickle cell disease (SCD) in adolescents and young adults (AYA): Transition from pediatric to adult care" and "Overview of the management and prognosis of sickle cell disease" and "Investigational therapies for sickle cell disease".)

OTHER DISEASE COMPLICATIONS

Hematuria and/or flank pain

Evaluation of hematuria and flank pain — Hematuria is one of the more common findings in SCD, occurring in approximately 6 percent of children and a large but unknown proportion of adults [101]. There are a number of possible causes ranging from benign to serious. Hematuria is also common in individuals with sickle cell trait, as discussed separately. (See "Sickle cell trait", section on 'Urologic and kidney disease'.)

All individuals with SCD (or sickle cell trait) who develop microscopic or macroscopic hematuria should be evaluated for the cause; this finding should not merely be attributed to the underlying disease. (See "Evaluation of gross hematuria in children" and "Etiology and evaluation of hematuria in adults".)

Common causes of hematuria and flank pain in SCD (and sickle cell trait) include renal papillary necrosis (RPN) and urinary tract infections (UTIs). As discussed below, renal medullary carcinoma (RMC) is a rare aggressive malignancy that should be considered in any individual with unexplained hematuria. (See 'Renal infarction and papillary necrosis' below and 'UTI and pyelonephritis' below and 'Renal medullary carcinoma' below.)

The evaluation of microscopic hematuria is discussed separately. (See "Evaluation of microscopic hematuria in children" and "Etiology and evaluation of hematuria in adults".)

For macroscopic (gross) hematuria, the evaluation includes urinalysis, culture, and typically kidney ultrasound to evaluate for papillary necrosis, kidney stones, and other kidney lesions. As with any individual who presents with bleeding, impaired hemostasis should be ruled out as a contributing factor with testing such as coagulation studies, platelet count, and other testing as indicated based on the type and severity of bleeding. (See "Approach to the child with bleeding symptoms" and "Approach to the adult with a suspected bleeding disorder".)

All individuals with SCD who have hematuria that does not resolve should undergo computed tomography (CT) scanning to evaluate for RMC, as the sensitivity of ultrasound for this malignancy is low and the only opportunity for cure is early detection. (See 'Renal medullary carcinoma' below and "Etiology and evaluation of hematuria in adults", section on 'Selection of modality'.)

Renal infarction and papillary necrosis — Infarction and RPN are common in sickle cell nephropathy (SCN), seen in as many as 23 to 40 percent of individuals [102,103]. Contributing factors include interstitial nephritis, acute pyelonephritis, obstructive uropathy, diabetes mellitus, or analgesic use.

The bleeding is typically mild, painless, and self-limited [8]. It may be complicated by urinary tract obstruction or infection [33,104]. Acute segmental or total renal infarcts may present with nausea, vomiting, flank or abdominal pain, fever, and hypertension (presumably renin-mediated) [105,106].

Renal infarction and RPN are generally diagnoses of exclusion but can be detected by imaging in many cases; other causes of hematuria and/or abdominal or flank pain need to be eliminated before these findings can be attributed to RPN.

Kidney ultrasound is a useful initial test; it typically shows increased echogenicity of the inner medulla and, in more advanced cases, a filling defect in the area of the medullary tip. Definitive diagnosis requires CT. CT may be omitted in selected individuals with symptoms that are milder or less concerning. Re-imaging is not required unless there are specific findings such as obstruction or the need for urologic intervention [107].

Management depends on the degree of hematuria:

For microscopic hematuria, hydration is often adequate treatment in the short term.

For macroscopic hematuria, bedrest and aggressive hydration are often used.

Bedrest is intended to avoid dislodging blood clots.

Hydration and alkalization of the urine may be used to reduce the toxicity from heme pigment, and diuretics may be used to increase the urinary flow rate. The rationale and details of therapy are discussed separately. (See "Prevention and treatment of heme pigment-induced acute kidney injury (including rhabdomyolysis)", section on 'Prevention'.)

Exchange blood transfusion may be used to lower the percentage of hemoglobin S in some cases. (See "Red blood cell transfusion in sickle cell disease: Indications and transfusion techniques", section on 'Exchange blood transfusion'.)

For severe or refractory hematuria, a number of other interventions have been described in case reports, although none of these have been studied in detail. These include:

Desmopressin (DDAVP) [108]

An antifibrinolytic agent (eg, tranexamic acid [TXA], epsilon-aminocaproic acid [EACA]) [99,109,110]

Embolization of the involved vessel or balloon tamponade [111]

Unilateral nephrectomy has been performed but is not recommended because bleeding can recur in the remaining kidney.

Antifibrinolytic therapy was previously reserved for individuals for whom other therapies have been ineffective, due to the risk of urinary obstruction from clots in the collecting system. However, we have used low-dose antifibrinolytic therapy in individuals with hematuria due to papillary necrosis and have found it to be safe and effective, as discussed below [109]. (See 'Clinical vignettes' below.)

Any individual with infarction or RPN significant enough to cause hematuria who is not already receiving hydroxyurea should discuss the potential benefits, which in most cases are likely to outweigh the risks.

UTI and pyelonephritis — Urinary tract infections (UTIs) and pyelonephritis are more common in individuals with SCD than those in the general population [104]. The mechanisms may include higher rates of volume depletion and urinary tract obstruction or inflammation related to infarction or papillary necrosis. These infections are especially concerning because individuals with SCD are functionally asplenic and at risk of sepsis from encapsulated organisms.

UTI or pyelonephritis may precipitate vaso-occlusive complications, and it is important to consider these (and other) infections in individuals presenting with pain, especially if atypical for their usual vaso-occlusive pain. (See "Evaluation of acute pain in sickle cell disease".)

This is particularly important in children, who often do not present with typical urinary tract symptoms such as urinary urgency, frequency, or dysuria, and in individuals with an indwelling urinary catheter. (See "Urinary tract infections in infants and children older than one month: Clinical features and diagnosis", section on 'Clinical presentation' and "Catheter-associated urinary tract infection in adults".)

These infections should also be suspected in an individual who presents with hematuria, dysuria, flank pain, abdominal pain, nausea/vomiting, and/or other systemic symptoms. Evaluation should include urinalysis with culture and sensitivities, imaging to assess for ureteral obstruction, as well as pregnancy testing for women of childbearing potential. (See "Acute infectious cystitis: Clinical features and diagnosis in children older than two years and adolescents" and "Acute complicated urinary tract infection (including pyelonephritis) in adults and adolescents".)

All individuals with SCD who present with fever should be treated promptly (within 60 minutes, preferably after cultures have been obtained). Individuals with SCD who have fever are often advised to have antibiotics available at home. Antibiotic regimens should include appropriate coverage for likely pathogens and encapsulated organisms, as discussed in separate topic reviews. (See "Clinical features, evaluation, and management of fever in patients with impaired splenic function", section on 'Evaluation and management'.)

Renal medullary carcinoma — RMC is a rare, highly aggressive non-clear-cell kidney cancer predominately found in people with sickle cell trait [112-116]. The typical onset is in early adulthood, and men are affected approximately twice as frequently as women [117]. Presenting findings include flank pain and hematuria; some individuals have a palpable mass, with right kidney involvement more common than left [113]. The disease is highly aggressive and often metastatic on presentation [114].

The biologic basis for the unique features of RMC have not been fully explained, but loss of expression of the chromatin remodeling factor SMARCB1 appears to be important [118,119].

RMC is not restricted to those with sickle cell trait; it can also occur in individuals with SCD, and clinicians should have a low threshold for considering the diagnosis in an individual with flank pain and/or hematuria [117,120]. RMC may appear as a cyst on ultrasound; this should be further evaluated with contrast-enhanced CT or magnetic resonance imaging (MRI) [121]. Scans for metastatic disease should be obtained in patients who are found to have RMC.

The optimal management of RMC is unknown. Nephrectomy is usually performed, and chemotherapy or targeted (biologic) therapy is often administered. The tumors appear to be platinum-sensitive [122-124]. Inhibitors of programmed cell death protein 1 (PD-1) such as pembrolizumab and nivolumab are under investigation, and enrollment in a clinical trial is encouraged [125]. (See "The treatment of advanced non-clear cell renal carcinoma", section on 'Collecting duct and renal medullary carcinoma'.)

The prognosis for RMC is poor. In a 2017 series of 52 individuals with RMC, 38 (73 percent) underwent nephrectomy [126]. Median survival was 16 months in those who had a nephrectomy and seven months in those who did not. Seven (13 percent) survived beyond two years.

Acute kidney injury — Acute kidney injury (AKI; eg, decline in urine output or rise in serum creatinine by ≥0.3 mg/dL developing within hours to days) occurs in as many as 17 percent of children and 10 percent of adults with SCD who present to the hospital [127,128].

In a Pediatric Health Information System database review that included pediatric admission for vaso-occlusive pain events, 2.5 percent of admissions were complicated by AKI [129]. AKI was associated with increased likelihood of admission to the intensive care unit, longer hospitalization, and increased likelihood of readmission.

In another study, the incidence of AKI during vaso-occlusive pain episodes was approximately 4 percent and appeared to be limited to those with acute chest syndrome and pulmonary hypertension, suggesting a potential role for right ventricular dysfunction and venous congestion [130].

Causes of AKI — There are a number of potential causes of AKI in SCD:

Hypovolemia due to dehydration, infection, or splenic sequestration

Rhabdomyolysis

Nephrotoxic medications

Renal vein thrombosis

Urinary tract obstruction (eg, due to blood clots)

Hepatorenal syndrome related to hepatic failure from iron overload

Individuals with SCD can also develop AKI for any of the reasons that apply to the general population. (See "Diagnostic approach to adult patients with subacute kidney injury in an outpatient setting" and "Evaluation of acute kidney injury among hospitalized adult patients" and "Acute kidney injury in children: Clinical features, etiology, evaluation, and diagnosis".)

Acute pain AKI is common during admission for acute pain events. One study observed AKI in 33 of 197 admissions for acute vaso-occlusive pain (17 percent) [131]. The likelihood of AKI also correlated with the extent of hemoglobin reduction in these patients. Both anemia and hemolysis are likely causal; free heme can be nephrotoxic. In a study in 185 children admitted for acute SCD pain, 61 (33 percent) had AKI on admission and 29 of 124 (23 percent) developed AKI [132].

The nonsteroidal antiinflammatory drug (NSAID) ketorolac is a known risk factor for AKI and further increases the risk of kidney damage during admissions for acute pain [131,132]. (See 'Other morbidities contributing to declining kidney function' above and "Acute vaso-occlusive pain management in sickle cell disease", section on 'Therapies we do not use'.)

Surgery – AKI may also complicate surgery, which is common in people with SCD. In a study involving patients with SCD undergoing total knee arthroplasty (TKA), the risk of AKI was double that of individuals without SCD undergoing TKA [133]. The cause of postoperative AKI is most likely multifactorial and appears to be correlated with postoperative thrombosis, acute chest syndrome, and anemia. These findings underscore the need to optimize perioperative management in all patients with SCD, especially those with underlying kidney disease.

COVID-19 – Individuals with SCD have an increased risk of AKI from COVID-19, which can lead to dialysis-dependence and death [134,135].

The risk of acute kidney disease may also be increased in individuals with sickle cell trait, especially those with preexisting chronic kidney disease (CKD) [136].

Management and prognosis of AKI — The prognosis for individual episodes of AKI appears to be favorable, with 10 of 12 individuals (83 percent) having recovery of kidney function in one series [127]. However, repeated episodes of acute or subacute kidney injury may contribute to the development of CKD. (See 'Pathogenesis' above.)

Evaluation and standard interventions for AKI are similar to the general population and include thorough history and physical examination, examination of the urine chemistry and sediment, and imaging of the kidneys (algorithm 1). Comprehensive discussions of the general approach are presented separately. (See "Acute kidney injury in children: Clinical features, etiology, evaluation, and diagnosis" and "Diagnostic approach to adult patients with subacute kidney injury in an outpatient setting" and "Evaluation of acute kidney injury among hospitalized adult patients".)

The same indications for dialysis in AKI are used in SCD as in individuals in the general population. (See "Kidney replacement therapy (dialysis) in acute kidney injury in adults: Indications, timing, and dialysis dose".)

There is no evidence to suggest that transfusion improves outcomes in individuals with SCD who develop AKI. A 2014 National Heart, Lung, and Blood Institute (NHLBI) guideline on the management of SCD made a consensus recommendation that transfusions not be used in AKI unless there are other indications (eg, symptomatic anemia, splenic sequestration, acute chest syndrome, acute stroke) [10,68]. These and other indications are discussed separately. (See "Red blood cell transfusion in sickle cell disease: Indications and transfusion techniques", section on 'Indications for transfusion'.)

Blood pressure abnormalities

Hypotension — Hypotension is more common in SCD than the general population [137]. Proposed mechanisms include prostaglandin-mediated changes in vascular tone and salt reabsorption [69,137]. Individuals with impaired urinary concentration (hyposthenuria) are at higher risk for volume depletion and may have less ability to compensate for a reduction in oral intake by increasing water retention in the kidney [10].

In patients whose baseline blood pressure readings are typically low, relatively "normal" blood pressure readings may actually represent significant hypertension, with attendant risks of adverse cardiovascular outcomes. Likewise, the use of antihypertensive agents to lower intraglomerular pressure may result in more severe hypotension than in the general population.

As a result, it is important to ensure adequate hydration, especially during acute pain episodes, acute chest syndrome, or infections, when oral intake may be decreased and insensible water losses may be increased. (See "Overview of the management and prognosis of sickle cell disease", section on 'Hydration'.)

Hypertension — The incidence of hypertension in SCD is markedly lower than in the general population of Black individuals (2 to 6 percent versus 28 percent). The challenge in making the diagnosis of hypertension in children and adults with SCD is that SCD is associated with a decrease in blood pressure [138-140]. Furthermore, higher systolic blood pressures (typically not considered in the hypertension range) are associated with earlier mortality, strokes, silent strokes, elevated tricuspid jet velocity, and CKD [140-144].

All individuals with SCD should be closely monitored for the development of relative hypertension and masked hypertension. The pulse pressure is emerging as an important factor for consideration as well. (See "Increased pulse pressure".)

Risk factors for hypertension such as nocturnal hypoxemia, which is common in the SCD population, as well as other risk factors seen in non-SCD populations (eg, high body mass index, high dietary sodium intake), should be sought and addressed appropriately. Sleep studies should be obtained in selected individuals with suspected nocturnal desaturation. (See "Overview of the clinical manifestations of sickle cell disease", section on 'Sleep disordered breathing and nocturnal hypoxemia' and "Overview of hypertension in adults", section on 'Risk factors for primary (essential) hypertension'.)

For those with hypertension, management involves nonpharmacologic interventions such as weight loss (if overweight), salt restriction (<100 mEq [2.3 g of sodium or 6 g of sodium chloride]/day), and exercise, similar to the non-SCD population. For those with reproducible hypertension, an antihypertensive medication is initiated with a goal blood pressure of ≤130/80. (See "Overview of hypertension in acute and chronic kidney disease" and "Antihypertensive therapy and progression of nondiabetic chronic kidney disease in adults".)

Antihypertensive medications should be used judiciously in individuals with SCD due to the risks of hypotension and volume depletion. For individuals with SCD who require antihypertensive medications, we usually use an angiotensin-converting enzyme (ACE) inhibitor or an angiotensin II receptor blocker (ARB). The rationale is that these agents reduce proteinuria and lessen the likelihood of progression to end-stage kidney disease (ESKD), as noted above. Individuals with peripheral or pulmonary edema are also treated with a loop diuretic or dialysis if indicated (indications the same as in the non-SCD population). Individuals without proteinuria may be treated with a renin-angiotensin or an ACE inhibitor, an ARB, or, if edema is present, a diuretic. The rationale for these choices and options for second- and third-line therapy are presented separately. (See 'Nephrologist referral and CKD interventions' above.)

Hyperuricemia and hyperkalemia — Hyperuricemia and gout may be an early sign of CKD with impaired uric acid excretion [10]. Joint pain from gout may be mistakenly attributed to vaso-occlusive pain. (See "Pathophysiology of gout", section on 'Hyperuricemia and gout' and "Evaluation of acute pain in sickle cell disease".)

Details of the evaluation and management of gout are presented separately. (See "Clinical manifestations and diagnosis of gout" and "Treatment of gout flares" and "Pharmacologic urate-lowering therapy and treatment of tophi in patients with gout".)

Hyperkalemia is often a secondary effect from untreated metabolic acidosis and possibly renal tubular acidosis in individuals with SCD. Hyperkalemia may also complicate acute or CKD. (See "Causes and evaluation of hyperkalemia in adults", section on 'Acute and chronic kidney disease'.)

Severe hemolysis can also cause hyperkalemia or contribute to hyperkalemia in individuals with underlying kidney disease (eg, after transfusion). The hyperkalemia in these cases is often transient and resolves with treatment of the underlying condition. It is also possible for hemolysis to occur at the time of the blood draw (eg, due to hemolysis in the collection needle), and unexpected increases in serum potassium should be confirmed by repeat testing with attention to minimizing hemolysis. (See "Causes and evaluation of hyperkalemia in adults", section on 'Pseudohyperkalemia'.)

For severe hyperkalemia (eg, with concern about arrhythmias), management is the same as in the non-SCD population, as discussed in separate topic reviews. (See "Management of hyperkalemia in children" and "Treatment and prevention of hyperkalemia in adults".)

Enuresis — Enuresis (nighttime bedwetting) is more common in children and young adults with SCD compared with age-matched controls, with prevalences as high as 9 percent [145-147]. A number of potential causes may contribute including hyposthenuria, sleep-disordered breathing, and psychological stress. (See 'Pathogenesis' above and "Overview of the pulmonary complications of sickle cell disease".)

Evaluation – Evaluation is similar to that in individuals without SCD; there should be a lower threshold for asking patients and parents about this symptom.

Treatment – The optimal treatment of nocturnal enuresis in SCD is unknown. Behavioral strategies with noninvasive alarms may be beneficial [146].

Children with SCD and enuresis have disrupted sleep patterns, and repetitive attempts to wake them up during the night may amplify their associated sleep disorder. The benefit of desmopressin in SCD is poorly studied, and it should not be the first-line therapy for enuresis as it could result in clinical hyponatremia. (See "Nocturnal enuresis in children: Etiology and evaluation" and "Nocturnal enuresis in children: Management" and "Nocturia: Clinical presentation, evaluation, and management in adults".)

Priapism and erectile dysfunction — Priapism (unwanted or sustained erection in the absence of sexual activity) is a known complication of SCD that does not appear to correlate with other urinary complications. Repeated episodes of priapism can lead to erectile dysfunction. The urology or nephrology evaluation is an appropriate time to ask about these symptoms, since some individuals may be reluctant to mention them to their primary clinician. Evaluation and management are discussed separately. (See "Priapism and erectile dysfunction in sickle cell disease".)

CLINICAL VIGNETTES — The following case vignettes illustrate the presentations of selected complications and our approach to evaluating and treating them:

Renal tubular acidosis – A 25-year-old male with SCD and a history of stroke was receiving chronic transfusion therapy for nine years, with a remarkable recovery from neurologic injury but significant iron overload related to transfusions. He had difficulty adhering to therapy with the iron chelator deferoxamine, which requires nightly continuous infusions for 10 to 12 hours. He switched to deferasirox (25 mg/kg/day) with close monitoring of ferritin and serum chemistries. (See 'Routine surveillance and early detection' above.)

His ferritin improved, but over a period of several months he developed bone pain, generalized weakness, and dehydration that were attributed to his underlying SCD. Subsequently, his serum creatinine increased mildly to 1.2 mg/dL and his urinalysis began to demonstrate proteinuria. Several months later he became confused and short of breath and was rushed to the emergency room, where he was found to have severe acidosis, proteinuria, and urinary potassium wasting (random urinary potassium of 45 mmol/L). He was diagnosed with severe renal tubular acidosis and treated with intravenous bicarbonate and correction of electrolyte abnormalities, as well as stopping the deferasirox, which is known to cause impaired kidney function and renal tubular acidosis. His laboratory parameters returned to normal; however, he continued to require daily potassium and phosphate supplementation. His iron chelation medication was changed to daily deferiprone, with deferoxamine on weekends.

Papillary necrosis – An 18-year-old African American male with Hb SC disease presented to the hospital with flank pain and macroscopic hematuria that was present on awakening from sleep [109]. He had mild thrombocytopenia attributed to hypersplenism (platelet count stable between 68,000 to 78,000/microL) and normal coagulation testing including testing for von Willebrand disease (VWD). Cystourethroscopy showed multiple clots in the bladder and bleeding from the left kidney; these were also present on CT scanning (with contrast). Ultrasound showed renal papillary necrosis, and CT showed no evidence of a kidney tumor. (See 'Evaluation of hematuria and flank pain' above.)

He was treated with intravenous hydration, bladder irrigation, and exchange transfusions, but he continued to have massive hematuria [109]. Nephrectomy was considered, but in an attempt to avoid nephrectomy, he was treated with low-dose oral epsilon aminocaproic acid (EACA; 50 mg/kg orally every eight hours). Treatment resulted in marked resolution of hematuria within a few hours; however, the creatinine rose from 1.3 to 1.8 mg/dL, leading to cessation of EACA. Kidney ultrasound showed no obstruction, and his urine became grossly bloody, so EACA was restarted at 20 mg/kg every 12 hours. This resulted in cessation of hematuria with a stable creatinine at 1.2 to 1.3 mg/dL. He was able to be discharged home on EACA at a dose of 5 mg/kg orally every eight hours, which he continued for approximately four weeks.

Renal medullary carcinoma – A 16-year-old African American male with SCD presented with several months of abdominal pain and microscopic hematuria [148]. CT scans showed a primary mass in the kidney with retroperitoneal lymphadenopathy and lesions in the lungs and liver. Biopsy showed renal medullary carcinoma. He was treated with neoadjuvant chemotherapy (carboplatin, gemcitabine, and bortezomib) followed by nephrectomy and palliative radiation for pain. He had a transient response followed by widespread metastatic disease and died 20 months after diagnosis.

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Chronic kidney disease in adults" and "Society guideline links: Sickle cell disease and thalassemias".)

PATIENT PERSPECTIVE TOPIC — Patient perspectives are provided for selected disorders to help clinicians better understand the patient experience and patient concerns. These narratives may offer insights into patient values and preferences not included in other UpToDate topics. (See "Patient perspective: Sickle cell disease".)

SUMMARY AND RECOMMENDATIONS

Mechanisms of kidney injury – Sickle cell disease (SCD) has several effects on the kidney, collectively termed "sickle cell nephropathy" (SCN). Kidney damage is multifactorial and begins in childhood but is often only appreciated retrospectively. By adulthood, one-third to one-fourth of individuals are affected, especially with Hb SS or sickle-beta0 thalassemia. (See 'Pathogenesis' above and 'Prevalence' above.)

Screening – Screening for SCN is essential for all individuals with SCD. Routine evaluations including blood pressure, urinalysis including albumin-to-creatinine ratio, and metabolic panel with estimation of glomerular filtration rate (eGFR). Testing is initiated between three to five years and no later than 10 years. Children are seen every four to six months and adults every two to three months. Caveats are noted above. (See 'Evaluation' above.)

PreventionHydroxyurea is the mainstay of treatment in SCD. For individuals with SCN hypertension who are not already taking hydroxyurea, we suggest hydroxyurea (Grade 2C). This is likely to reduce proteinuria and delay or prevent the progression of chronic kidney disease (CKD). Other approaches are under investigation. (See 'Role of hydroxyurea and transfusions' above and "Hydroxyurea use in sickle cell disease" and 'Investigational approaches' above.)

Therapy for CKD – For hypertension and/or urine protein excretion >500 mg per day, we recommend adding an angiotensin-converting enzyme (ACE) inhibitor or an angiotensin II receptor blocker (ARB), which is likely to slow CKD progression and lower blood pressure. (See "Antihypertensive therapy and progression of nondiabetic chronic kidney disease in adults", section on 'Summary and recommendations'.)

We feel more strongly about using an ACE inhibitor or ARB if there is hypertension or urine protein ≥1000 mg per day (approximately equivalent to a urine protein to creatinine ratio [uPCR] >100 mg/mmol [>884 mg/g]). Individuals with SCD generally have lower blood pressures, and attention to dosing is needed to avoid hypotension. (See 'Nephrologist referral and CKD interventions' above and 'Hypertension' above.)

Dialysis and transplant – Individuals with SCD should have timely access to preventive therapies and dialysis and/or kidney transplantation when needed. An erythropoiesis-stimulating agent (ESA) may be used for worsening anemia, with caveats regarding the target hemoglobin level (to avoid hyperviscosity) and avoidance of routine iron administration, as most individuals with SCD have iron overload. (See 'Nephrologist referral and CKD interventions' above and 'ESKD interventions (dialysis and transplant)' above and 'Prognosis' above.)

Hematuria – Hematuria is common and often due to renal papillary necrosis and/or infection. Renal medullary carcinoma is rare but should be considered. (See 'Hematuria and/or flank pain' above.)

AKI – Acute kidney injury (AKI) is common and may be due to hypovolemia, rhabdomyolysis, nephrotoxic medications (including nonsteroidal antiinflammatory drugs [NSAIDs], particularly ketorolac), urinary tract obstruction, and others. Management is similar to individuals without SCD. (See 'Acute kidney injury' above.)

Hypotension – Hypotension is more common and hypertension less common in SCD compared with the general population. Attention should be paid to hydration and judicious use of antihypertensive therapies as indicated. (See 'Blood pressure abnormalities' above.)

Metabolic and urologic problems – Other findings include hyperuricemia, hyperkalemia, enuresis, priapism, or erectile dysfunction. (See 'Hyperuricemia and hyperkalemia' above and 'Enuresis' above and "Priapism and erectile dysfunction in sickle cell disease".)

ACKNOWLEDGMENT — UpToDate gratefully acknowledges Stanley L Schrier, MD (deceased), who contributed as Section Editor on earlier versions of this topic and was a founding Editor-in-Chief for UpToDate in Hematology.

  1. Ataga KI, Derebail VK, Archer DR. The glomerulopathy of sickle cell disease. Am J Hematol 2014; 89:907.
  2. Vichinsky E. Chronic organ failure in adult sickle cell disease. Hematology Am Soc Hematol Educ Program 2017; 2017:435.
  3. Sharpe CC, Thein SL. How I treat renal complications in sickle cell disease. Blood 2014; 123:3720.
  4. Naik RP, Derebail VK. The spectrum of sickle hemoglobin-related nephropathy: from sickle cell disease to sickle trait. Expert Rev Hematol 2017; 10:1087.
  5. Haymann JP, Stankovic K, Levy P, et al. Glomerular hyperfiltration in adult sickle cell anemia: a frequent hemolysis associated feature. Clin J Am Soc Nephrol 2010; 5:756.
  6. Nath KA, Hebbel RP. Sickle cell disease: renal manifestations and mechanisms. Nat Rev Nephrol 2015; 11:161.
  7. Boyle SM, Jacobs B, Sayani FA, Hoffman B. Management of the Dialysis Patient with Sickle Cell Disease. Semin Dial 2016; 29:62.
  8. Pham PT, Pham PC, Wilkinson AH, Lew SQ. Renal abnormalities in sickle cell disease. Kidney Int 2000; 57:1.
  9. Hatch FE, Culbertson JW, Diggs LW. Nature of the renal concentrating defect in sickle cell disease. J Clin Invest 1967; 46:336.
  10. https://www.nhlbi.nih.gov/health-topics/evidence-based-management-sickle-cell-disease.
  11. Ataga KI, Saraf SL, Derebail VK. The nephropathy of sickle cell trait and sickle cell disease. Nat Rev Nephrol 2022; 18:361.
  12. Sundaram N, Bennett M, Wilhelm J, et al. Biomarkers for early detection of sickle nephropathy. Am J Hematol 2011; 86:559.
  13. Tharaux PL, Hagège I, Placier S, et al. Urinary endothelin-1 as a marker of renal damage in sickle cell disease. Nephrol Dial Transplant 2005; 20:2408.
  14. Heimlich JB, Speed JS, O'Connor PM, et al. Endothelin-1 contributes to the progression of renal injury in sickle cell disease via reactive oxygen species. Br J Pharmacol 2016; 173:386.
  15. Taori KB, Chaudhary RS, Attarde V, et al. Renal Doppler indices in sickle cell disease: early radiologic predictors of renovascular changes. AJR Am J Roentgenol 2008; 191:239.
  16. Xu JZ, Garrett ME, Soldano KL, et al. Clinical and metabolomic risk factors associated with rapid renal function decline in sickle cell disease. Am J Hematol 2018; 93:1451.
  17. Gurkan S, Scarponi KJ, Hotchkiss H, et al. Lactate dehydrogenase as a predictor of kidney involvement in patients with sickle cell anemia. Pediatr Nephrol 2010; 25:2123.
  18. Youssry I, Makar S, Fawzy R, et al. Novel marker for the detection of sickle cell nephropathy: soluble FMS-like tyrosine kinase-1 (sFLT-1). Pediatr Nephrol 2015; 30:2163.
  19. Saraf SL, Zhang X, Kanias T, et al. Haemoglobinuria is associated with chronic kidney disease and its progression in patients with sickle cell anaemia. Br J Haematol 2014; 164:729.
  20. Mohtat D, Thomas R, Du Z, et al. Urinary transforming growth factor beta-1 as a marker of renal dysfunction in sickle cell disease. Pediatr Nephrol 2011; 26:275.
  21. Ghobrial EE, Abdel-Aziz HA, Kaddah AM, Mubarak NA. Urinary Transforming Growth Factor β-1 as a Marker of Renal Dysfunction in Sickle Cell Disease. Pediatr Neonatol 2016; 57:174.
  22. Bergmann S, Zheng D, Barredo J, et al. Renal kallikrein: a risk marker for nephropathy in children with sickle cell disease. J Pediatr Hematol Oncol 2006; 28:147.
  23. Adly AA, Ismail EA, Andrawes NG, et al. Soluble Fas/FasL ratio as a marker of vasculopathy in children and adolescents with sickle cell disease. Cytokine 2016; 79:52.
  24. Voskaridou E, Terpos E, Michail S, et al. Early markers of renal dysfunction in patients with sickle cell/beta-thalassemia. Kidney Int 2006; 69:2037.
  25. Alvarez O, Lopez-Mitnik G, Zilleruelo G. Short-term follow-up of patients with sickle cell disease and albuminuria. Pediatr Blood Cancer 2008; 50:1236.
  26. Ashley-Koch AE, Okocha EC, Garrett ME, et al. MYH9 and APOL1 are both associated with sickle cell disease nephropathy. Br J Haematol 2011; 155:386.
  27. Nolan VG, Ma Q, Cohen HT, et al. Estimated glomerular filtration rate in sickle cell anemia is associated with polymorphisms of bone morphogenetic protein receptor 1B. Am J Hematol 2007; 82:179.
  28. Saraf SL, Zhang X, Shah B, et al. Genetic variants and cell-free hemoglobin processing in sickle cell nephropathy. Haematologica 2015; 100:1275.
  29. Schaefer BA, Flanagan JM, Alvarez OA, et al. Genetic Modifiers of White Blood Cell Count, Albuminuria and Glomerular Filtration Rate in Children with Sickle Cell Anemia. PLoS One 2016; 11:e0164364.
  30. Garrett ME, Soldano KL, Erwin KN, et al. Genome-wide meta-analysis identifies new candidate genes for sickle cell disease nephropathy. Blood Adv 2023; 7:4782.
  31. Saborio P, Scheinman JI. Sickle cell nephropathy. J Am Soc Nephrol 1999; 10:187.
  32. Drawz P, Ayyappan S, Nouraie M, et al. Kidney Disease among Patients with Sickle Cell Disease, Hemoglobin SS and SC. Clin J Am Soc Nephrol 2016; 11:207.
  33. Wesson DE. The initiation and progression of sickle cell nephropathy. Kidney Int 2002; 61:2277.
  34. de Jong PE, Statius van Eps LW. Sickle cell nephropathy: new insights into its pathophysiology. Kidney Int 1985; 27:711.
  35. Bhathena DB, Sondheimer JH. The glomerulopathy of homozygous sickle hemoglobin (SS) disease: morphology and pathogenesis. J Am Soc Nephrol 1991; 1:1241.
  36. Lebensburger JD, Aban I, Pernell B, et al. Hyperfiltration during early childhood precedes albuminuria in pediatric sickle cell nephropathy. Am J Hematol 2019; 94:417.
  37. Kasztan M, Fox BM, Lebensburger JD, et al. Hyperfiltration predicts long-term renal outcomes in humanized sickle cell mice. Blood Adv 2019; 3:1460.
  38. Schmitt F, Martinez F, Brillet G, et al. Early glomerular dysfunction in patients with sickle cell anemia. Am J Kidney Dis 1998; 32:208.
  39. Ter Maaten JC, Gans ROB, De Jong PE. Comprehensive Clinical Nephrology, Johnson RJ, Feehally J (Eds), Elsevier, 2003. p.665.
  40. Aygun B, Mortier NA, Smeltzer MP, et al. Hydroxyurea treatment decreases glomerular hyperfiltration in children with sickle cell anemia. Am J Hematol 2013; 88:116.
  41. Ware RE, Rees RC, Sarnaik SA, et al. Renal function in infants with sickle cell anemia: baseline data from the BABY HUG trial. J Pediatr 2010; 156:66.
  42. Marsenic O, Couloures KG, Wiley JM. Proteinuria in children with sickle cell disease. Nephrol Dial Transplant 2008; 23:715.
  43. Allon M. Renal abnormalities in sickle cell disease. Arch Intern Med 1990; 150:501.
  44. De Jong PE, de Jong-van Den Berg LT, Statius van Eps LW. The tubular reabsorption of phosphate in sickle-cell nephropathy. Clin Sci Mol Med 1978; 55:429.
  45. Amarapurkar P, Roberts L, Navarrete J, El Rassi F. Sickle Cell Disease and Kidney. Adv Chronic Kidney Dis 2022; 29:141.
  46. Bayazit AK, Noyan A, Aldudak B, et al. Renal function in children with sickle cell anemia. Clin Nephrol 2002; 57:127.
  47. Batlle D, Itsarayoungyuen K, Arruda JA, Kurtzman NA. Hyperkalemic hyperchloremic metabolic acidosis in sickle cell hemoglobinopathies. Am J Med 1982; 72:188.
  48. Silva Junior GB, Vieira AP, Couto Bem AX, et al. Renal tubular dysfunction in sickle cell disease. Kidney Blood Press Res 2013; 38:1.
  49. Nasr SH, Markowitz GS, Sentman RL, D'Agati VD. Sickle cell disease, nephrotic syndrome, and renal failure. Kidney Int 2006; 69:1276.
  50. Serjeant GR. Sickle-cell disease. Lancet 1997; 350:725.
  51. Bakir AA, Hathiwala SC, Ainis H, et al. Prognosis of the nephrotic syndrome in sickle glomerulopathy. A retrospective study. Am J Nephrol 1987; 7:110.
  52. Falk RJ, Scheinman J, Phillips G, et al. Prevalence and pathologic features of sickle cell nephropathy and response to inhibition of angiotensin-converting enzyme. N Engl J Med 1992; 326:910.
  53. Powars DR, Elliott-Mills DD, Chan L, et al. Chronic renal failure in sickle cell disease: risk factors, clinical course, and mortality. Ann Intern Med 1991; 115:614.
  54. Maigne G, Ferlicot S, Galacteros F, et al. Glomerular lesions in patients with sickle cell disease. Medicine (Baltimore) 2010; 89:18.
  55. Gaartman AE, van Tuijn CFJ, Nur E, et al. The association between renal function decline and disease severity in sickle cell disease. Am J Hematol 2023; 98:E95.
  56. Gosmanova EO, Zaidi S, Wan JY, Adams-Graves PE. Prevalence and progression of chronic kidney disease in adult patients with sickle cell disease. J Investig Med 2014; 62:804.
  57. Guasch A, Navarrete J, Nass K, Zayas CF. Glomerular involvement in adults with sickle cell hemoglobinopathies: Prevalence and clinical correlates of progressive renal failure. J Am Soc Nephrol 2006; 17:2228.
  58. Kruzel-Davila E, Wasser WG, Skorecki K. APOL1 Nephropathy: A Population Genetics and Evolutionary Medicine Detective Story. Semin Nephrol 2017; 37:490.
  59. Kormann R, Jannot AS, Narjoz C, et al. Roles of APOL1 G1 and G2 variants in sickle cell disease patients: kidney is the main target. Br J Haematol 2017; 179:323.
  60. Naik RP, Irvin MR, Judd S, et al. Sickle Cell Trait and the Risk of ESRD in Blacks. J Am Soc Nephrol 2017; 28:2180.
  61. Winer JC, Yee ME, Ataga KI, et al. Patients with sickle cell disease who develop end-stage kidney disease continue to experience poor survival - A 19-year United States Renal Data System study. Br J Haematol 2022; 199:e43.
  62. Yeruva SL, Paul Y, Oneal P, Nouraie M. Renal Failure in Sickle Cell Disease: Prevalence, Predictors of Disease, Mortality and Effect on Length of Hospital Stay. Hemoglobin 2016; 40:295.
  63. Belisário AR, Simões E Silva AC, Moura ICG, et al. Estimated glomerular filtration rate in Brazilian adults with sickle cell disease: results from the REDS-III multicenter cohort study. Ann Hematol 2023; 102:1019.
  64. Shatat IF, Qanungo S, Hudson S, et al. Changes in Urine Microalbumin-to-Creatinine Ratio in Children with Sickle Cell Disease over Time. Front Pediatr 2016; 4:106.
  65. Bharani A, Manchanda R, Singh RK, Prashant S. Distal renal tubular acidosis in sickle cell anemia. Saudi J Kidney Dis Transpl 2018; 29:1000.
  66. Papadopoulos N, Vasiliki A, Aloizos G, et al. Hyperchloremic metabolic acidosis due to deferasirox in a patient with beta thalassemia major. Ann Pharmacother 2010; 44:219.
  67. Dee CM, Cheuk DK, Ha SY, et al. Incidence of deferasirox-associated renal tubular dysfunction in children and young adults with beta-thalassaemia. Br J Haematol 2014; 167:434.
  68. Yawn BP, Buchanan GR, Afenyi-Annan AN, et al. Management of sickle cell disease: summary of the 2014 evidence-based report by expert panel members. JAMA 2014; 312:1033.
  69. Allon M, Lawson L, Eckman JR, et al. Effects of nonsteroidal antiinflammatory drugs on renal function in sickle cell anemia. Kidney Int 1988; 34:500.
  70. Asnani MR, Lynch O, Reid ME. Determining glomerular filtration rate in homozygous sickle cell disease: utility of serum creatinine based estimating equations. PLoS One 2013; 8:e69922.
  71. Asnani M, Reid M. Cystatin C: a useful marker of glomerulopathy in sickle cell disease? Blood Cells Mol Dis 2015; 54:65.
  72. Mendoza SA. Nephrotoxic drugs. Pediatr Nephrol 1988; 2:466.
  73. Alvarez O, Miller ST, Wang WC, et al. Effect of hydroxyurea treatment on renal function parameters: results from the multi-center placebo-controlled BABY HUG clinical trial for infants with sickle cell anemia. Pediatr Blood Cancer 2012; 59:668.
  74. Laurin LP, Nachman PH, Desai PC, et al. Hydroxyurea is associated with lower prevalence of albuminuria in adults with sickle cell disease. Nephrol Dial Transplant 2014; 29:1211.
  75. Bartolucci P, Habibi A, Stehlé T, et al. Six Months of Hydroxyurea Reduces Albuminuria in Patients with Sickle Cell Disease. J Am Soc Nephrol 2016; 27:1847.
  76. Fitzhugh CD, Wigfall DR, Ware RE. Enalapril and hydroxyurea therapy for children with sickle nephropathy. Pediatr Blood Cancer 2005; 45:982.
  77. Becker AM. Sickle cell nephropathy: challenging the conventional wisdom. Pediatr Nephrol 2011; 26:2099.
  78. https://www.abbottpointofcare.com/download?docUri=/technical-library/static-assets/technical-documentation/714183-00Y.pdf (Accessed on November 04, 2016).
  79. Alvarez O, Nottage K, Simpson LM, et al. Kidney function of transfused children with sickle cell anemia: Baseline data from the TWiTCH study with comparison to non-transfused cohorts. Am J Hematol 2017; 92:E637.
  80. Alvarez O, Montane B, Lopez G, et al. Early blood transfusions protect against microalbuminuria in children with sickle cell disease. Pediatr Blood Cancer 2006; 47:71.
  81. Sasongko TH, Nagalla S, Ballas SK. Angiotensin-converting enzyme (ACE) inhibitors for proteinuria and microalbuminuria in people with sickle cell disease. Cochrane Database Syst Rev 2013; :CD009191.
  82. Foucan L, Bourhis V, Bangou J, et al. A randomized trial of captopril for microalbuminuria in normotensive adults with sickle cell anemia. Am J Med 1998; 104:339.
  83. Aoki RY, Saad ST. Enalapril reduces the albuminuria of patients with sickle cell disease. Am J Med 1995; 98:432.
  84. Thrower A, Ciccone EJ, Maitra P, et al. Effect of renin-angiotensin-aldosterone system blocking agents on progression of glomerulopathy in sickle cell disease. Br J Haematol 2019; 184:246.
  85. Olaniran KO, Eneanya ND, Zhao SH, et al. Mortality and Hospitalizations among Sickle Cell Disease Patients with End-Stage Kidney Disease Initiating Dialysis. Am J Nephrol 2020; 51:995.
  86. Bynum JP, Zachary A, Ness PM, et al. Transfusion of leukoreduced blood products and risk of antibody-mediated rejection of renal allografts. Transfusion 2018; 58:1951.
  87. Abbott KC, Hypolite IO, Agodoa LY. Sickle cell nephropathy at end-stage renal disease in the United States: patient characteristics and survival. Clin Nephrol 2002; 58:9.
  88. Wong WY, Elliott-Mills D, Powars D. Renal failure in sickle cell anemia. Hematol Oncol Clin North Am 1996; 10:1321.
  89. Nath J, McDaid J, Bentall A, et al. Sickle cell and renal transplant: a national survey and literature review. Exp Clin Transplant 2012; 10:1.
  90. Sharpe CC, Suddle A, Stuart-Smith S. An Overview of Solid Organ Transplantation in Patients With Sickle Cell Disease. Transplantation 2023; 107:596.
  91. Scheinman JI. Sickle cell disease and the kidney. Nat Clin Pract Nephrol 2009; 5:78.
  92. Huang E, Parke C, Mehrnia A, et al. Improved survival among sickle cell kidney transplant recipients in the recent era. Nephrol Dial Transplant 2013; 28:1039.
  93. Ojo AO, Govaerts TC, Schmouder RL, et al. Renal transplantation in end-stage sickle cell nephropathy. Transplantation 1999; 67:291.
  94. Okafor UH, Aneke E. Outcome and challenges of kidney transplant in patients with sickle cell disease. J Transplant 2013; 2013:614610.
  95. Montgomery R, Zibari G, Hill GS, Ratner LE. Renal transplantation in patients with sickle cell nephropathy. Transplantation 1994; 58:618.
  96. Miner DJ, Jorkasky DK, Perloff LJ, et al. Recurrent sickle cell nephropathy in a transplanted kidney. Am J Kidney Dis 1987; 10:306.
  97. Sherwood JB, Goldwasser E, Chilcote R, et al. Sickle cell anemia patients have low erythropoietin levels for their degree of anemia. Blood 1986; 67:46.
  98. Han J, Zhou J, Kondragunta V, et al. Erythropoiesis-stimulating agents in sickle cell anaemia. Br J Haematol 2018; 182:602.
  99. Hariri E, Mansour A, El Alam A, et al. Sickle cell nephropathy: an update on pathophysiology, diagnosis, and treatment. Int Urol Nephrol 2018; 50:1075.
  100. Han J, Molokie RE, Hussain F, et al. Voxelotor and albuminuria in adults with sickle cell anaemia. Br J Haematol 2022; 197:e63.
  101. Stallworth JR, Tripathi A, Jerrell JM. Prevalence, treatment, and outcomes of renal conditions in pediatric sickle cell disease. South Med J 2011; 104:752.
  102. Odita JC, Ugbodaga CI, Okafor LA, et al. Urographic changes in homozygous sickle cell disease. Diagn Imaging 1983; 52:259.
  103. McCall IW, Moule N, Desai P, Serjeant GR. Urographic findings in homozygous sickle cell disease. Radiology 1978; 126:99.
  104. Bruno D, Wigfall DR, Zimmerman SA, et al. Genitourinary complications of sickle cell disease. J Urol 2001; 166:803.
  105. Domanovits H, Paulis M, Nikfardjam M, et al. Acute renal infarction. Clinical characteristics of 17 patients. Medicine (Baltimore) 1999; 78:386.
  106. Lumerman JH, Hom D, Eiley D, Smith AD. Heightened suspicion and rapid evaluation with CT for early diagnosis of partial renal infarction. J Endourol 1999; 13:209.
  107. Henderickx MMEL, Brits T, De Baets K, et al. Renal papillary necrosis in patients with sickle cell disease: How to recognize this 'forgotten' diagnosis. J Pediatr Urol 2017; 13:250.
  108. Moudgil A, Kamil ES. Protracted, gross hematuria in sickle cell trait: response to multiple doses of 1-desamino-8-D-arginine vasopressin. Pediatr Nephrol 1996; 10:210.
  109. Gabrovsky A, Aderinto A, Spevak M, et al. Low dose, oral epsilon aminocaproic acid for renal papillary necrosis and massive hemorrhage in hemoglobin SC disease. Pediatr Blood Cancer 2010; 54:148.
  110. Black WD, Hatch FE, Acchiardo S. Aminocaproic acid in prolonged hematuria of patients with sicklemia. Arch Intern Med 1976; 136:678.
  111. Herard A, Colin J, Youinou Y, et al. Massive gross hematuria in a sickle cell trait patient with renal papillary necrosis. Conservative approach using a balloon ureteral catheter to tamponade the papilla bleeding. Eur Urol 1998; 34:161.
  112. Avery RA, Harris JE, Davis CJ Jr, et al. Renal medullary carcinoma: clinical and therapeutic aspects of a newly described tumor. Cancer 1996; 78:128.
  113. Hakimi AA, Koi PT, Milhoua PM, et al. Renal medullary carcinoma: the Bronx experience. Urology 2007; 70:878.
  114. Davis CJ Jr, Mostofi FK, Sesterhenn IA. Renal medullary carcinoma. The seventh sickle cell nephropathy. Am J Surg Pathol 1995; 19:1.
  115. Shetty A, Matrana MR. Renal medullary carcinoma: a case report and brief review of the literature. Ochsner J 2014; 14:270.
  116. Çalışkan S, Gökçe AM, Gümrükçü G, Önenerk M. Renal Medullary Carcinoma; A Rare Entity. Iran J Med Sci 2017; 42:215.
  117. Alvarez O, Rodriguez MM, Jordan L, Sarnaik S. Renal medullary carcinoma and sickle cell trait: A systematic review. Pediatr Blood Cancer 2015; 62:1694.
  118. Msaouel P, Tannir NM, Walker CL. A Model Linking Sickle Cell Hemoglobinopathies and SMARCB1 Loss in Renal Medullary Carcinoma. Clin Cancer Res 2018; 24:2044.
  119. Swartz MA, Karth J, Schneider DT, et al. Renal medullary carcinoma: clinical, pathologic, immunohistochemical, and genetic analysis with pathogenetic implications. Urology 2002; 60:1083.
  120. Alvarez OA. Renal Medullary Carcinoma: The Kidney Cancer That Affects Individuals With Sickle Cell Trait and Disease. J Oncol Pract 2017; 13:424.
  121. Greco F, Faiella E, Santucci D, et al. Imaging of Renal Medullary Carcinoma. J Kidney Cancer VHL 2017; 4:1.
  122. Strouse JJ, Spevak M, Mack AK, et al. Significant responses to platinum-based chemotherapy in renal medullary carcinoma. Pediatr Blood Cancer 2005; 44:407.
  123. Walsh A, Kelly DR, Vaid YN, et al. Complete response to carboplatin, gemcitabine, and paclitaxel in a patient with advanced metastatic renal medullary carcinoma. Pediatr Blood Cancer 2010; 55:1217.
  124. Maroja Silvino MC, Venchiarutti Moniz CM, Munhoz Piotto GH, et al. Renal medullary carcinoma response to chemotherapy: a referral center experience in Brazil. Rare Tumors 2013; 5:e44.
  125. Beckermann KE, Jolly PC, Kim JY, et al. Clinical and immunologic correlates of response to PD-1 blockade in a patient with metastatic renal medullary carcinoma. J Immunother Cancer 2017; 5:1.
  126. Shah AY, Karam JA, Malouf GG, et al. Management and outcomes of patients with renal medullary carcinoma: a multicentre collaborative study. BJU Int 2017; 120:782.
  127. Sklar AH, Perez JC, Harp RJ, Caruana RJ. Acute renal failure in sickle cell anemia. Int J Artif Organs 1990; 13:347.
  128. Mammen C, Bissonnette ML, Matsell DG. Acute kidney injury in children with sickle cell disease-compounding a chronic problem. Pediatr Nephrol 2017; 32:1287.
  129. McCormick M, Richardson T, Warady BA, et al. Acute kidney injury in paediatric patients with sickle cell disease is associated with increased morbidity and resource utilization. Br J Haematol 2020; 189:559.
  130. Audard V, Homs S, Habibi A, et al. Acute kidney injury in sickle patients with painful crisis or acute chest syndrome and its relation to pulmonary hypertension. Nephrol Dial Transplant 2010; 25:2524.
  131. Baddam S, Aban I, Hilliard L, et al. Acute kidney injury during a pediatric sickle cell vaso-occlusive pain crisis. Pediatr Nephrol 2017.
  132. Batte A, Menon S, Ssenkusu J, et al. Acute kidney injury in hospitalized children with sickle cell anemia. BMC Nephrol 2022; 23:110.
  133. Viswanathan VK, Ramanan SP, Beale J, et al. How does sickle cell disease affect the peri-operative outcome in patients undergoing total knee arthroplasty? A large-scale, National Inpatient Sample-based study. Arch Orthop Trauma Surg 2023; 143:5261.
  134. Minniti CP, Zaidi AU, Nouraie M, et al. Clinical predictors of poor outcomes in patients with sickle cell disease and COVID-19 infection. Blood Adv 2021; 5:207.
  135. Fisher LA, Asnani M. Acute kidney injury in Jamaicans with sickle cell disease hospitalized with COVID-19 infection. EJHaem 2023; 4:37.
  136. Verma A, Huffman JE, Gao L, et al. Association of Kidney Comorbidities and Acute Kidney Failure With Unfavorable Outcomes After COVID-19 in Individuals With the Sickle Cell Trait. JAMA Intern Med 2022; 182:796.
  137. Hatch FE, Crowe LR, Miles DE, et al. Altered vascular reactivity in sickle hemoglobinopathy. A possible protective factor from hypertension. Am J Hypertens 1989; 2:2.
  138. Aderibigbe A, Omotoso AB, Awobusuyi JO, Akande TM. Arterial blood pressure in adult Nigerian sickle cell anaemia patients. West Afr J Med 1999; 18:114.
  139. Johnson CS, Giorgio AJ. Arterial blood pressure in adults with sickle cell disease. Arch Intern Med 1981; 141:891.
  140. Pegelow CH, Colangelo L, Steinberg M, et al. Natural history of blood pressure in sickle cell disease: risks for stroke and death associated with relative hypertension in sickle cell anemia. Am J Med 1997; 102:171.
  141. Rodgers GP, Walker EC, Podgor MJ. Is "relative" hypertension a risk factor for vaso-occlusive complications in sickle cell disease? Am J Med Sci 1993; 305:150.
  142. Ohene-Frempong K, Weiner SJ, Sleeper LA, et al. Cerebrovascular accidents in sickle cell disease: rates and risk factors. Blood 1998; 91:288.
  143. DeBaun MR, Sarnaik SA, Rodeghier MJ, et al. Associated risk factors for silent cerebral infarcts in sickle cell anemia: low baseline hemoglobin, sex, and relative high systolic blood pressure. Blood 2012; 119:3684.
  144. Gordeuk VR, Sachdev V, Taylor JG, et al. Relative systemic hypertension in patients with sickle cell disease is associated with risk of pulmonary hypertension and renal insufficiency. Am J Hematol 2008; 83:15.
  145. Esezobor CI, Akintan P, Nwaogazie U, et al. Enuresis in children and adolescents with sickle cell anaemia is more frequent and substantially different from the general population. PLoS One 2018; 13:e0201860.
  146. Wolf RB, Kassim AA, Goodpaster RL, DeBaun MR. Nocturnal enuresis in sickle cell disease. Expert Rev Hematol 2014; 7:245.
  147. Ekinci O, Celik T, Ünal Ş, et al. Nocturnal enuresis in sickle cell disease and thalassemia major: associated factors in a clinical sample. Int J Hematol 2013; 98:430.
  148. Marsh A, Golden C, Hoppe C, et al. Renal medullary carcinoma in an adolescent with sickle cell anemia. Pediatr Blood Cancer 2014; 61:567.
Topic 7199 Version 41.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟