Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

87 Renal Insufficiency and Ischemic Nephropathy

Joshua Augustine, MD, Alvin C. Wee, MD, Venkatesh Krishnamurthi, MD, and David A. Goldfarb, MD

ACUTE KIDNEY INJURY Such fluctuation in kidney function can create challenges in drug
dosing in the hospital setting (Lewis et al., 2016). In addition, drug
Acute kidney injury (AKI) in is defined by an acute rise in serum dosing has historically relied on outdated methods of measurement
creatinine (SCr) and/or an acute decline in urine output. This of kidney function, such as SCr alone or the calculation of creatinine
abrupt decline in kidney function occurs over hours to days and clearance using formulas such as Cockcroft-Gault. The Cockcroft-Gault
results in the accumulation of byproducts of metabolism and the formula uses patient weight but does not account for morbid obesity,
dysregulation of electrolyte homeostasis, acid/base, and volume which may positively skew the estimation of creatinine clearance
status. Changes in blood urea nitrogen (BUN) and SCr have been well above the true GFR (Bouquegneau et al., 2016).
used as surrogate markers for changes in kidney function. BUN can Another method occasionally used to estimate kidney function
be altered by other factors and may be disproportionately elevated in the hospital setting is a 24-hour urine collection for creatinine
in states of volume depletion, in hypercatabolic states, or in marked clearance. Twenty-four–hour urine is often imprecise and may result
increased in protein loads seen with gastrointestinal bleeding or in errors related to over- or undercollection. It may be used to assess
total parenteral nutrition. Creatinine is a metabolite of muscle and kidney function in a patient who is suspected of having impaired
is used to calculate the estimated glomerular filtration rate (GFR), kidney function beyond that reflected by the SCr alone, or to screen
using GFR equations that also account for age, gender, and black for renal recovery in a patient with nonoliguric AKI. However, in
race. The two equations most commonly used and often reported addition to errors in collection, the percentage of creatinine excreted
with routine lab values include the Modification of Diet in Renal in the tubules can vary and tends to be a higher percentage of total
Disease (MDRD) and the Chronic Kidney Disease Epidemiology creatinine excretion at lower levels of GFR. Tubular secretion of
Collaboration (CKD-EPI) equations (Levey et al., 1999, 2009). The creatinine may account for more than a third of creatinine measured
CKD-EPI equation is a modification of the MDRD formula and is in the urine and thus may artificially inflate creatinine clearance
less likely to underestimate true GFR in patients with early stages relative to true GFR (Doolan et al., 1962).
of chronic kidney disease (CKD) (Levey et al., 2009). It is more The gold standard for evaluating kidney function is a measure-
predictive of mortality associated with CKD than the MDRD equation ment of GFR using a marker of pure glomerular filtration (Stevens
(Matsushita et al., 2012). et al., 2009). Inulin has been used historically but is no longer
Because of its dependence on muscle mass, SCr may overestimate available in the United States. GFR is more often measured by nuclear
kidney function in cases of muscle wasting, such as in patients with medicine testing using iothalamate or other isotopes. Few hospitals
cirrhosis or prolonged intensive care unit (ICU) stays (Schetz et al., routinely use nuclear measurement of GFR, but it has been a valuable
2014; Skluzacek et al., 2003). Alternatively, at extremes of high muscle tool particularly in research designed to derive estimation formulas.
mass, SCr may underestimate kidney function. Another marker of It has also been used in some kidney transplant centers to evaluate
kidney function that has been compared with SCr is the measurement kidney function in living kidney donor candidates.
of cystatin C, a low-molecular-weight protein that is a member of One set of criteria used to define AKI is known as the Risk, Injury
the cystatin superfamily of cysteine protease inhibitors (Newman Failure, Loss of kidney function, and End-stage kidney disease (RIFLE)
et al., 1995). Unlike creatinine, cystatin is not secreted by renal criteria (Bellomo et al., 2004; Table 87.1), which divides AKI and
tubules, although it does undergo renal tubular metabolism. Cystatin chronic kidney failure into stages based on severity and duration.
C does not appear to be as strongly correlated with muscle mass The Kidney Disease: Improving Global Outcomes (KDIGO) definition
and thus may improve the accuracy of estimating kidney function of AKI is also widely used and is described in Table 87.2. Contrasting
in patients with extremes of muscle mass and nutrition, such as from the RIFLE criteria, KDIGO includes an acute rise in SCr more
patients with cirrhosis (Pöge et al., 2006). However, cystatin C levels than 0.3 mg/dL within 48 hours as a definition of AKI, along with
appear to be altered in states of inflammation, thyroid disease, and a rise in SCr of at least 1.5 times baseline within 7 days, or urine
smoking, making it an imperfect replacement for SCr (Knight et al., volume below 0.5 mL/kg/h for 6 consecutive hours (KDIGO, 2012).
2004; Manetti et al., 2005). Combining formulas that estimate GFR Unlike RIFLE, the KDIGO criteria do not include scoring related to
using both SCr and cystatin C leads to improved precision and prolonged kidney failure or end-stage renal disease (ESRD).
accuracy compared with using either variable alone (Inker et al., Use of these AKI definitions does not account for the cause of
2012). Online calculators that allow for combining of such formulas AKI, which may be rapidly correctable in cases of volume depletion,
are available. See The National Kidney Foundation website at https:// or more severe and sustained in cases of intrinsic ischemic kidney
www.kidney.org/professionals/KDOQI/gfr_calculator. injury. Therefore staging alone has some prognostic capability but
It is also important to recognize that changes in SCr may lag in should optimally be subdivided into cause of AKI. In addition, it is
appearance after an episode of AKI. For example, contrast nephropathy, not always clear whether a patient had a normal baseline SCr before
which may induce kidney injury immediately after contrast exposure, AKI, or developed “acute on chronic” kidney injury with underlying
is often associated in a change in SCr evident only after 24 to 48 CKD. Patients with CKD have a greater risk for AKI (Chawla et al.,
hours (Rich et al., 1990). For this reason, it is inappropriate to 2012). AKI may develop before a hospital admission, with an eleva-
estimate GFR using a single SCr value in a hospitalized patient tion in SCr on presentation, thus affecting definitions that use the
whose SCr is fluctuating. For example, a patient whose SCr doubles change in SCr from admission to a later hospital date. Regardless, it
from 1 to 2 mg/dL in 24 hours may have advanced kidney failure is recommended that the practitioner identify AKI early in its course
with lower clearance than a patient with a stable SCr of 2 mg/dL. because even small changes in SCr may indicate a significant change
Conversely, a patient recovering from AKI or after kidney transplanta- in kidney function. Early consultation with nephrology may also
tion with an SCr falling from 6 mg/dL to 3 mg/dL in 24 hours has improve outcomes in hospitalized patients with AKI (Balasubramanian
a better GFR than would be reflected by a stable SCr of 3 mg/dL. et al., 2011).

1921
Downloaded for FK UMI Makassar (mahasiswafkumi09@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier on January 10, 2021.
For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
1922 PART VIII Renal Physiology and Pathophysiology

TABLE 87.1 The Risk, Injury Failure, Loss of Kidney TABLE 87.3 Cause of Hospital-Acquired Acute Kidney
Function, and End-Stage Kidney Disease Injury in 748 Patients in Thirteen Tertiary
(RIFLE) Criteria for Acute Kidney Injury Care Centers in Madrid, Spain

RIFLE CRITERIA: STAGES CAUSE PERCENTAGE

Stage I Increase in serum creatinine 50%–99% OR Acute tubular necrosis 45


Urine output < 0.5 mL/kg/h for 6–12 h Prerenal disease 21
Stage 2 Increase in serum creatinine of 100%–200% OR Acute on chronic kidney disease (primarily 13
Urine output < 0.5 mL/kg/h for 12–24 h resulting from acute tubular necrosis or
prerenal)
Failure Increase in serum creatinine ≥ 200% OR
Obstructive disease 10
Increase in serum creatinine > 0.5 mg/dL to > Glomerulonephritis or vasculitis 4
4 mg/dL OR Acute interstitial nephritis 2
Urine output < 0.3 mL/kg/h for >24 h or anuria for Atheroembolic disease 1
>12 h OR
Initiation of renal replacement therapy
Loss Need for renal replacement therapy for >4 weeks
End stage Need for renal replacement therapy for >3 months The kidney can maintain normal renal blood flow and GFR with
systemic perfusion pressures as low as 55 to 60 mm Hg under normal
conditions (Walsh et al., 2013), although such autoregulation of
kidney blood flow may be compromised in CKD, making patients
with chronic disease more susceptible to AKI. In the setting of low
TABLE 87.2 The Kidney Disease: Improving Global systemic blood pressure, renal angiotensin II, which has selective
Outcomes (KDIGO) Definition of Acute vasoconstriction on the efferent (postglomerular) arteriole, and
Kidney Injury vasodilatory prostaglandins, which dilate the afferent (preglomerular)
arteriole, play an important role in maintaining glomerular hydrostatic
KDIGO CRITERIA: STAGES pressure and GFR. Drugs that inhibit angiotensin, such as
angiotensin-converting enzyme (ACE) inhibitors and angiotensin
Stage 1 Increase in serum creatinine ≥ 0.3 mg/dL or
receptor blockers (ARBs), exert a vasodilatory effect on the efferent
50%–99% OR arteriole and thus reduce glomerular pressure and filtration frac-
Urine output < 0.5 mL/kg/h for 6–12 h tion. Such agents are of great value in treatment of diabetic
Stage 2 Increase in serum creatinine of 100%–200% OR nephropathy and congestive heart failure and help to reduce
Urine output < 0.5 mL/kg/h for 12–24 h proteinuria and convey renal protection in proteinuric kidney
disease. However, they may increase the risk of prerenal AKI as
Stage 3 Increase in serum creatinine ≥ 200% OR well as hyperkalemia during periods of hypotension and hypo-
Increase in serum creatinine ≥ 0.3 mg/dL to ≥ perfusion and are thus often held in the hospital setting after
4 mg/dL OR identification of AKI. In addition, drugs that inhibit prostaglandins
Urine output < 0.3 mL/kg/h for > 24 h or anuria for such as nonsteroidal anti-inflammatory drugs (NSAIDs) can contribute
≥ 12 h OR to the risk of prerenal and intrinsic AKI (Whelton, 1999).
Initiation of renal replacement therapy Although prerenal AKI may often occur as a result of volume
In patients < 18 y/o: a decline in estimated depletion such as after hemorrhage or GI losses, it can also occur
in instances of total body overload. For example, congestive heart
glomerular filtration rate < 35 mL/min/1.73 m2
failure and cirrhosis often occur with total body overload, edema
and/or ascites and yet are defined by prerenal AKI typically with
a low urine sodium concentration and low FeNa. Other causes of
prerenal AKI related to volume overload include third spacing related
A classic way to divide AKI is by “prerenal,” “intrinsic,” and to low albumin states, sepsis, and burn injury. These conditions can
“postrenal” or obstructive causes. However, prolonged prerenal create a reduction in “effective” circulating volume. Such prerenal
physiology may lead to intrinsic kidney ischemia and injury, such states are difficult to manage because administration of crystalloid
as is seen with prolonged hepatorenal syndrome (HRS). Prolonged only worsens total body volume overload and third spacing. Treatment
obstruction can also lead to intrinsic kidney damage. The greatest is directed toward treating the underlying disease states in an
proportion of hospital acquired AKI is secondary to acute tubular effort to optimize systemic hemodynamics and renal perfusion.
necrosis (ATN). A Spanish analysis of 748 cases of AKI in 13 tertiary In heart failure, paradoxically, prerenal AKI may improve with diuresis
care hospital centers revealed that ATN accounted for 45% of all using agents such as loop diuretics. Improvement in right heart
AKI (Liaño et al., 1996). Alternatively, immune-mediated AKI from function (Testani et al., 2010) and reduction in venous back pressure
glomerulonephritis or acute interstitial nephritis accounted for less on the kidneys (Bock and Gottlieb, 2010) may explain improved
than 10% of cases (Table 87.3). kidney function with diuresis in heart failure.
The sympathetic nervous system and the renin-angiotensin cascade
Prerenal Kidney Injury are activated in prerenal AKI in the face of reduced blood flow.
In addition, hypotension is a powerful stimulus for the release of
Prerenal AKI is caused by new renal hypoperfusion that leads to a antidiuretic hormone (ADH) from the posterior pituitary gland.
fall in GFR typically with urinary sodium avidity, as noted by a ADH release is triggered by baroceptor activation in response to low
reduction in urinary sodium or the fractional excretion of sodium systemic blood pressure, and it causes free water retention even in the
(FeNa) in the urine. The hallmark of prerenal disease is its revers- face of normal or low serum sodium concentrations (Bankir et al.,
ibility after treatment of the underlying cause, with absence of 2017). ADH binds to V2 receptors in the distal collecting tubule
structural damage to the kidney if treatment promptly. Recovery of the kidney, leading to increased transcription and insertion of
of renal function within 24 to 72 hours of volume resuscitation or aquaporin-2 water channels and free water absorption, often leading
correction of renal hemodynamics helps to define prerenal AKI in to hyponatremia. The most common cause of hyponatremia in the
isolation. hospital setting is related to such “hypovolemic” hyponatremia, as

Downloaded for FK UMI Makassar (mahasiswafkumi09@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier on January 10, 2021.
For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Chapter 87 Renal Insufficiency and Ischemic Nephropathy 1923

opposed to other conditions such as the syndrome of inappropriate ATN, bilateral vascular occlusion, hemolytic-uremic syndrome, or
ADH secretion (SIADH). It is important to assess volume status severe cases of crescentic glomerulonephritis such as anti-glomerular
through measurements of urine sodium, FeNa in the urine, and basement membrane (GBM) disease.
other measures such as orthostatic blood pressure monitoring. In A kidney ultrasound is a sensitive test for identifying hydronephro-
hyponatremia related to volume depletion, urinary sodium, and sis related to obstruction. However, there is a high false-positive rate,
FeNa tend to be low (UNa < 20 mEq/L, FeNa < 1%). Serum sodium which can vary depending upon the index of suspicion (Kamholtz
levels correct quickly with volume repletion, typically with the use of et al., 1989). The negative predictive value is high, although false-
normal saline or other isotonic crystalloid therapy. Hyponatremia in negative tests may also be seen with early obstruction. In addition,
patients with volume overload but ineffective circulating volume is metastatic malignancies involving the ureters or retroperitoneal
also typically associated with low urine sodium. It is more difficult fibrosis may mask hydronephrosis by restricting the ability of
to manage and often requires diuresis and free water restriction. the ureters to expand (Somerville et al., 1992). Further imaging
Tolvaptan, an arginine vasopressin receptor antagonist, blocks the may be indicated, such as computed tomography (CT) scanning,
V2 receptor and leads to a rapid water diuresis in patients with a with noncontrast imaging if there is suspicion or concern for kidney
high ADH response. It has been used in patients with SIADH and stones. A simple bladder scan may reveal a distended bladder resulting
in patients with congestive heart failure (Konstam et al., 2007). The from bladder outlet obstruction. Foley catheter placement relieves
use of tolvaptan has been curtailed somewhat by the rapid correction obstruction related to bladder outlet obstruction and allows for
of serum sodium that can be seen, high cost of the agent, and a sampling of the urine for urinalysis (UA) and culture. Dilation at
“black box” warning against its use in patients with liver impairment, the level of the ureteropelvic junction with minimal bladder filling is
leading to avoidance in cirrhotic patients. more typically a result of a kidney stone or ureteropelvic malignancy.
HRS represents a unique and severe form of prerenal AKI in One challenge that urologists commonly face after the relief of
patients with cirrhosis, acute hepatitis, or any acute form of hepatic obstructive uropathy is “postobstructive diuresis.” This usually
decompensation. HRS can be seen in the hospital setting, and the represents an appropriate response to prolonged obstruction with
risk increases with additional known risk factors, including bacterial elimination of excess fluid and sodium. However, there may be
peritonitis and gastrointestinal bleeding (Wu et al., 2006). Dilation dysregulation with inappropriate water loss resulting from tubular
of the splanchnic vasculature mediated by nitric oxide appears to injury or an osmotic effect created by excretion of elevated blood
drive HRS pathophysiology (Iwakiri, 2007). Temporary improvement urea nitrogen (Schlossberg and Vaughan, 1984). This is a condition
in AKI associated with HRS has been associated with the use of that typically lasts 24 to 72 hours and can result in dehydration and
albumin, along with midodrine, an α1 agonist, which mediates hypernatremia, as well as electrolyte wasting. Generally it is recom-
vasoconstriction, and octreotide, a somatostatin analogue that mended to replace one-half of the urine output using hypotonic
decreases splanchnic blood flow (Angeli et al., 1999). Terlipressin, solutions, such as one-half normal saline and to monitor electrolytes
a vasopressin analogue, has been shown to improve renal function closely during this interval. Intravenous (IV) replacement with free
in HRS relative to midodrine and octreotide (Cavallin et al., 2015), water in dextrose may be appropriate in hypernatremic patients.
but it can cause intestinal ischemia and is currently not available in
the United States. The gold standard therapy for HRS is liver trans-
plantation, which can result in reversal of HRS pathophysiology and Intrinsic Kidney Injury
recovery of kidney function (Gonwa et al., 1991). Prolonged HRS, Acute Tubular Necrosis Resulting From Ischemic Injury
however, may result in permanent kidney injury, and the decision
to allocate a kidney allograft with a liver (simultaneous liver kidney Overall, AKI may occur in 2% to 7% of hospitalized patients in
transplantation) is in part related to the duration of AKI related to tertiary care centers, and the incidence in surgical or medical ICUs
HRS. Recent guidelines for combined liver kidney allocation define may exceed 25% to 35%. The type of AKI in hospitalized patients
advanced AKI with an eGFR of 25 mL/min or less and/or dialysis is ATN in the majority of cases (Myers and Moran, 1986; Uchino
dependence for 6 consecutive weeks as an appropriate duration of et al., 2005). Hypoperfusion and ischemic injury most commonly
AKI to warrant a combined liver and kidney transplant rather than cause ATN in the hospitalized setting related to operative bleeding
liver transplant alone (Formica et al., 2016). and intraoperative hypotension, sepsis, and shock.

Postrenal Kidney Injury Contrast Nephropathy


Obstruction of the urinary tract can be a cause of AKI and can Contrast-induced tubular injury (contrast nephropathy) is common
be acute or chronic, complete or partial, and involve any area of in the hospital setting. It is a type of ATN that is generally reversible
the lower urinary tract. Obstructive uropathy often occurs in the but can contribute to patient morbidity, mortality, and long-term
outpatient setting, and patients often are seen in the clinic or hos- kidney impairment. Patients who develop contrast nephropathy are
pital with functional impairment (Gottlieb et al., 1997). Unilateral more likely to be older with more comorbidities, including diabetes.
obstruction may initially go unnoticed in terms of kidney function Despite general reversibility of AKI, one analysis found a higher
and changes in SCr, although patients may gradually lose function 30-day mortality rate in patients with contrast nephropathy than those
in a single kidney, rendering them with reduced kidney mass and without after controlling for other factors (Giacoppo et al., 2015).
GFR. Chronic obstruction, whether partial or complete, may lead Iodinated contrast, particularly with higher osmolarity, has been
to tubule-interstitial atrophy and fibrosis, with irreversible loss of shown to induce renal vasoconstriction and medullary ischemia and
kidney function (Klahr, 1991). Patients may develop a distal renal also cause direct toxic effects at the proximal renal tubules (Persson
tubular acidosis, with hyperkalemia, mild sodium wasting, and et al., 2005). Contrast nephropathy may occur with a low FeNa (<1%)
reduced serum bicarbonate levels (Batlle and Arruda, 2018). The because of constrictive or renal tubular obstructive effects (Schwab
duration of obstruction predicts recovery of kidney function, and et al., 1989). An increase in SCr is generally observed at 24 to 48
ideally obstruction should be relieved as soon as possible and within hours after contrast exposure, and the AKI is typically nonoliguric.
a week of onset at most (Better et al., 1973). Muddy brown casts may be seen on urinary sediment.
Bilateral obstruction presents dramatically, often with oligoanuria, Risk factors for contrast nephropathy include older age, preexist-
advanced kidney failure, and electrolyte perturbations. Common ing CKD, diabetic nephropathy, congestive heart failure, hemo-
causes of bilateral obstruction include congenital posterior urethral dynamic instability, high osmolar contrast (Lautin et al., 1991),
valves in a younger population, bladder dysfunction including NSAID usage, higher volume of contrast, and volume depletion
neurogenic bladder, and prostatic disease in men. Oligoanuria before contrast exposure. Preventive measures should be used in
is a diagnostic sign that suggests complete obstruction and is an patients with CKD. NSAIDs, which can induce vasoconstriction,
uncommon feature of AKI overall. Other causes of anuric AKI should be avoided for 24 hours before contrast exposure. Generally,
include shock, which is readily apparent upon presentation, severe diuretics are held before the procedure as well to avoid volume

Downloaded for FK UMI Makassar (mahasiswafkumi09@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier on January 10, 2021.
For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
1924 PART VIII Renal Physiology and Pathophysiology

depletion. The use of isotonic IV fluids before and after contrast function. As stated earlier, aggressive volume resuscitation may prevent
is a staple in the prevention of contrast nephropathy despite only or curtail kidney injury.
limited data on protection (Jurado-Román et al., 2015; Luo et al.,
2014). There are various strategies related to IV hydration, but in Tumor Lysis Syndrome
general normal saline is given either in bolus form at 3 mL/kg for
an hour or 1 mL/kg for 6 to 12 hours before contrast exposure, Tumor lysis syndrome (TLS) is a condition seen in the presence of
followed by 1 to 1.5 mL/kg for 4 to 12 hours after contrast exposure. high tumor burden that may cause AKI in part because of precipitation
Sodium bicarbonate solutions may be used, although studies have of uric acid crystals in renal tubules causing urate nephropathy. TLS
been mixed in terms of any additional benefit over normal saline. is most commonly seen after treatment for high-grade lymphomas
Outcomes with the use of N-acetylcysteine have also been mixed. and leukemias but has also been reported in cases of solid tumors,
One large randomized trial with a 2×2 design was unable to dem- including renal cell and transitional cell carcinomas (Lin et al., 2007;
onstrate benefit with either sodium bicarbonate or N-acetylcysteine Norberg et al., 2014). Prophylactic use of agents such as rasburicase, a
compared to standard normal saline administration (Weisbord et al., urate-oxidase enzyme that converts uric acid to allantoin, has greatly
2018). Iso-osmolar contrast agents (e.g., iodixanol) are preferred in reduced the incidence of urate nephropathy associated with TLS.
patients with preexisting kidney disease, with slight benefit seen in Hyperphosphatemia with subsequent hypocalcemia and calcium-
one recent meta-analysis (Eng et al., 2016). Limiting contrast volume phosphate deposition remain features of TLS and may contribute
to the lowest amount possible is recommended in high risk patients, to AKI by causing nephrocalcinosis. Renal replacement therapy may
and diagnostic angiography may often be performed with minimal be required if the calcium-phosphorus product (Ca × Phos) exceeds
contrast (10–20 mL). 70 mg2/dL2 (Coiffier et al., 2008; Howard et al., 2011). Potassium levels
One important differential diagnosis in patients with AKI after may also become critically high. Cardiac monitoring and frequent
arterial instrumentation is atheroembolic kidney injury. Athero- assessment of electrolytes are essential in the management of TLS.
embolic disease should be suspected in patients who have other
evidence of embolic injury such as lesions in fingers or toes (e.g., Drug Toxicity as a Cause of Acute Kidney Injury
“blue toe syndrome”) or symptoms of gut ischemia such as pain,
nausea, and vomiting. Patients may develop transient eosinophilia Nephrotoxic pharmacologic agents are a common source of kidney
(Scolari et al., 2007) and hypocomplementemia (Lye et al., 1993). injury through direct toxic effects or by causing inflammation (intersti-
The natural history is different from contrast nephropathy, in that tial nephritis). Drugs and agents known to cause direct tubular toxicity
kidney injury may be gradual and unremitting, exhibiting a stuttering are shown in Table 87.4. Some, like aminoglycosides, amphotericin B,
pattern and progressing to advanced kidney failure. On biopsy, cisplatin, and ifosfamide, cause renal proximal tubular toxicity, which
needle-shaped clefts from cholesterol emboli that dissolve during is cumulative with repeated dosing. Patients with proximal tubular
fixation may be seen. These clefts are often surrounded by inflam- toxicity may initially have a “Fanconi syndrome,” with evidence
matory cells, including eosinophils. of proximal tubular wasting, including acidosis with an alkaline
urine (proximal renal tubular acidosis), phosphorus wasting, and
Pigment-Related Kidney Injury renal glycosuria.

Pigment injury related to hemoglobin or myoglobin may be


suspected in the appropriate clinical situation, such as severe TABLE 87.4 Drugs/Agents Known to Cause Direct
hemolysis or muscle breakdown related to trauma, toxicity, or Renal Tubular and Interstitial Toxicity
surgical injury, causing rhabdomyolysis. Muscle breakdown is
associated with elevation in creatine phosphokinase (CPK) in the Antibacterial/antifungal/antiviral
bloodstream along with high levels of phosphorus, potassium, and Aminoglycosides
uric acid related to cellular breakdown. Metabolic acidosis is common
Amphotericin B
and hypocalcemia can be severe related to cellular entry, and phos-
phorus binding is often seen. Myoglobin may cause positive testing Polymyxin, colistin
for blood by urine dipstick testing in the absence of true hematuria. Vancomycin
Myoglobinuria may also be seen early in the course and can contribute Acyclovir
to the production of red-brown–tinged urine. The risk of AKI is Foscarnet
related to the severity of muscle injury as well as volume status. Cidofovir
Early aggressive fluid resuscitation may prevent or limit the extent Indinavir
of kidney injury after muscle injury. Forced alkaline diuresis with Ritonavir
bicarbonate based therapy is controversial and requires close monitor- Tenofovir
ing of arterial pH and serum calcium levels every 2 hours ideally in Pentamidine
an ICU setting. Bicarbonate therapy may worsen hypocalcemia and
Iodinated contrast
promote calcium-phosphorus precipitation.
In urology, two specific clinical circumstances have been identified Nonsteroidal anti-inflammatory drugs
in association with rhabdomyolysis. The first relates to protracted Chemotherapeutic agents
exaggerated lithotomy positioning, as used in urethral stricture surgery Cisplatin
(Anema et al., 2000; Vijay et al., 2011) and robotic-assisted radical Carboplatin
prostatectomy (Keene et al., 2010). Gluteal muscles are often affected, Ifosfamide
and long exposure to lithotomy positioning greater than 5 hours Gemcitabine
appears to convey greater risk for muscle injury. Attention to padding, Methotrexate
positioning, and any maneuver that can reduce the duration of Immunosuppressive/immunomodulating therapy
exaggerated positioning help prevent this complication. A second Cyclosporine
type of surgery that has been associated with rhabdomyolysis is
Tacrolimus
laparoscopic nephrectomy, including after living kidney donor surgery
(Deane et al., 2008; Glassman et al., 2007; Kuang et al., 2002). Intravenous immunoglobulin
Prolonged lateral decubitus positioning appears to be a risk factor Psychotropics
for downside iliopsoas muscle injury, and risk factors also include Lithium
high body mass index. Unusually severe muscle pain early after Mannitol
surgery along with darkened red-brown urine should prompt an Zoledronic acid
early evaluation with blood CPK levels and measurement of kidney

Downloaded for FK UMI Makassar (mahasiswafkumi09@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier on January 10, 2021.
For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Chapter 87 Renal Insufficiency and Ischemic Nephropathy 1925

Some examples of agents recently identified as causing nephrotoxic- TABLE 87.5 Drugs Known to Cause Acute
ity include a number of chemotherapeutic agents that have shown Interstitial Nephritis
great promise in treating and stabilizing various metastatic malignan-
cies. For example, tyrosine kinase inhibitors, including sorafenib Antibiotics
and sunitinib, inhibit vascular endothelial growth factor (VEGF), Penicillins
and are known to cause hypertension, proteinuria (typically subne- Cephalosporins
phrotic), and AKI in certain patients. Inhibition of VEGF has been Sulfonamides
shown to predispose animal models to thrombotic microangiopathy
Ciprofloxacin
(TMA) (Eremina et al., 2008), and TMA has been a finding on kidney
biopsy in patients with tyrosine kinase inhibitor–related kidney injury Rifampin
(Bollée et al., 2009). Tyrosine kinase inhibitors are approved for Nonsteroidal anti-inflammatory drugs
the treatment of metastatic renal cell carcinoma, and urologists Including cyclooxygenase-2 inhibitors
involved with treatment should be aware of the potential neph- Proton pump inhibitors
rotoxicity associated with such agents. Checkpoint inhibitor chemotherapeutic agents
Another newer group of chemotherapeutic agents that may Allopurinol
induce AKI are the so-called checkpoint inhibitors, which are Loop and thiazide diuretics
immunomodulatory agents designed to enhance the immune Anticonvulsants
response against various malignancies. One class, known as pro- Phenytoin
grammed cell death receptor-1 (PD-1) inhibitors, includes medications
Carbamazepine
such as nivolumab and pembrolizumab. These agents are used in
metastatic renal cell and urothelial carcinomas, along with other Phenobarbital
malignancies. PD-1 inhibitors may cause immune mediated side effects Indinavir
including skin rash, colitis, hepatotoxicity, and pneumonitis. They Cimetidine
are also known to cause AKI with acute interstitial nephritis (AIN) Mesalamine
and occasionally immune complex glomerular disease manifesting
with heavy proteinuria (Cortazar et al., 2016; Izzedine et al., 2017).
AKI associated with these agents may require cessation of drug
therapy and/or corticosteroid treatment. infiltration (Saeki et al., 2010). Patients with kidney involvement
Recent epidemiologic data have also linked proton pump inhibitors often have extrarenal manifestations. Serum IgG-4 levels are elevated
(PPIs) to acute and chronic kidney disease. PPIs have long been in the majority of patients, and serum complement levels are often
known to cause AIN in a small percentage of patients (Geevasinga low, suggesting complement activation. Treatment with corticosteroid
et al., 2006). More recently, studies of large patient populations have therapy and other immunosuppressive agents has been proven effective
linked PPIs to CKD and ESRD (Lazarus et al., 2016; Xie et al., 2016). (Khosroshahi et al., 2015).
Duration of PPI usage appeared to correlate with risk. It is not entirely
clear whether such studies are flawed by residual confounding, but Renal Vein Thrombosis
there may be incidences of subacute AIN that go undetected in some
patients. It appears prudent to limit PPI usage in patients who can Renal vein thrombosis is a condition occasionally seen in nephrotic
otherwise be managed with dietary changes and/or the use of patients and patients with malignancy. Renal vein thrombosis is
histamine-2 receptor antagonists. Alternatively, patients with a history most commonly associated with membranous glomerulonephritis
of peptic ulcer or esophageal ulceration would likely retain benefit and is more common with glomerular disease associated with
that exceeds risk with maintenance PPI therapy. malignancy. Loss of anticoagulation factors such as anti-thrombin
In general, interstitial nephritis is common in the hospital III and an increase in procoagulant factors predispose nephrotic
setting and can occur acutely (AIN), often related to antibiotic patients to venous clotting (Loscalzo, 2013). The classic clinical
therapy, or chronically, often related to the long-term use of findings include flank pain (usually unilateral), microscopic or
NSAIDs. Table 87.5 lists common drugs associated with AIN. The gross hematuria, elevated lactate dehydrogenase (LDH), and an
acute presentation is classically described as including a rash, fever, enlarged, swollen kidney on imaging. Severe AKI is seen in cases
and eosinophilia. However, a report on AIN in 128 patients described of bilateral acute renal vein thrombosis.
a rash in only 15%, fever in 27%, and eosinophilia in only 23%, Urologists may see renal vein thrombosis associated with renal
and these findings were less common in AIN related to NSAIDs or cell carcinoma with renal vein involvement or external compression
PPIs (Baker and Pusey, 2004). Urinary eosinophils are commonly or postoperative thrombosis related to manipulation of the renal
measured but are neither sensitive nor specific for AIN (Muriithi circulation (partial nephrectomy or renal transplantation). Catheter-
et al., 2013). The most common urinary findings are pyuria and directed local thrombolytic therapy has been used with success in
hematuria, although a bland urine sediment does not exclude AIN. cases of AKI (Kim et al., 2006) and in renal transplantation (Fulton
Thus the diagnosis of AIN requires a high index of suspicion. Sterile et al., 2011). In cases of renal malignancy, nephrectomy with removal
pyuria and white cell casts on urine sediment support the diagnosis, of the tumor thrombus is usually the therapy of choice.
and confirmation is made by kidney biopsy. A Spanish registry analysis
found AIN in 12.9% of those biopsied for AKI, and the prevalence Clinical Approach to the Differential Diagnosis
of AIN appeared to be increasing in the more recent era, particularly of Acute Kidney Injury
in elderly patients (Goicoechea et al., 2013).
Non–drug-related AIN may be seen in various infections and Distinguishing among prerenal, intrinsic, and postrenal causes
rheumatologic diseases, including systemic lupus, sarcoidosis, and of AKI should be feasible with appropriate attention to the history,
Sjögren syndrome. Another emerging disease entity that may cause physical, laboratory, and imaging results. A thorough history and
AIN is so-called IgG-4 related disease, characterized by an inflam- physical examination to assess volume status, cardiovascular hemo-
matory infiltration predominantly containing IgG-4–positive dynamics, potential nephrotoxic insults, and evidence of systemic
plasma cells. Manifestations may include pancreatitis, lymph- disease should be undertaken in patients with AKI. All interventions
adenopathy, salivary or orbital involvement, cholangitis, and involve- and drug therapies surrounding an AKI event should be outlined
ment of other organ systems. Patients may have AKI related to against the timeline of changes in kidney function. Risk factors
retroperitoneal fibrosis, which can mimic the appearance of a associated with AKI such as advanced age, heart failure, liver failure,
tumor and cause obstructive uropathy. IgG-4–related disease may baseline CKD, diabetes, and recent surgery should be identified.
also cause intrinsic kidney injury with a dense AIN displaying a Exposures including radiocontrast and potential inciting pharma-
nodular pattern of inflammation on biopsy that can mimic tumor cologic agents may offer clues as to the cause. In perioperative AKI,

Downloaded for FK UMI Makassar (mahasiswafkumi09@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier on January 10, 2021.
For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
1926 PART VIII Renal Physiology and Pathophysiology

the duration of the procedure, blood loss, hemodynamic instability, improved sensitivity in identifying kidney stones relative to plain
integrity of the urinary tract, and intraoperative drug treatment are abdominal radiography and avoids the contrast required for IV
critical to review. urography (Pfister et al., 2003).
Vital signs and hemodynamic parameters should be assessed,
including the measurement of orthostatic hypotension, correctly Management of Acute Kidney Injury
performed, which may identify volume depletion and prerenal AKI.
Hypertension and third spacing of volume identify patients who The management of AKI focuses on reversing any prerenal condi-
would benefit from diuresis or possible renal replacement therapy. tion, restoring and maintaining euvolemia, and eliminating
Daily weights and accurate intake and output are essential in the nephrotoxic drugs when clinically appropriate. Early consultation
management of AKI in hospitalized patients. Central venous pressure with a nephrologist improves the outcome of AKI in some studies
monitoring may also be beneficial, particularly after major surgery of critically ill patients (Mehta et al., 2002; Ponce et al., 2011). During
and in an ICU setting. the initial evaluation, it is imperative to search for reversible causes
Examination of the UA and urinary studies is fundamental to such as volume depletion, obstruction, and vascular occlusion. A
the evaluation of the patient with AKI (Perazella and Parikh, 2009). trial of parenteral hydration with isotonic fluids may correct AKI
A basic UA may distinguish between various causes of AKI. A urine secondary to prerenal causes (Prowle et al., 2014). Thereafter fluid
sediment should also be examined. The nephrologist may assist in status should be monitored vigilantly to maintain euvolemia. In
identifying cells and casts after urine cytospin because relying on patients with oliguria, special attention must be provided to avoid
automated laboratory reporting of urinary casts from UA samples excessive hydration and volume overload, which could precipitate
is unreliable (Cavanaugh and Perazella, 2018). Heavy proteinuria, the need for renal replacement therapy.
hematuria, and red cell casts on urinary sediment are classic findings Diuretic therapy is commonly used in patients with AKI and
of acute glomerulonephritis, which may require a prompt biopsy volume overload. Response to diuretic therapy may depend on
and institution of immunosuppressive therapy. Proteinuria can further the severity of kidney injury, but diuretic therapy used appropriately
be quantified by a spot urinary protein to creatinine or albumin to neither hinders nor hastens recovery from AKI. Response to diuretic
creatinine ratio or by 24-hour urine collection. Nephrotic range therapy is a favorable prognostic sign in AKI (Cosentino, 1995).
proteinuria (>3.5 g protein/24 h) is highly suggestive of glomerular When dosing loop diuretics such as furosemide, a response to an
disease and typically prompts a biopsy, unless there is a high suspicion IV bolus can be assessed within 60 minutes of administration. If
for diabetic nephropathy or a contraindication to biopsy. Alternatively there is no significant augmentation in urine output, the dose should
and more commonly in the hospital setting, a bland urine is noted be increased to find the threshold dose for a given patient. If a dose
on UA, and “muddy brown” casts may be seen on urine microscopy, response is established, IV furosemide may be dosed every 6 to 8
helping to confirm the diagnosis of ATN. Sterile pyuria and hematuria hours at the amount required to elicit a diuretic response. Alternatively,
in association with white blood cell casts on urine microscopy is a continuous infusion may be administered. For example, if a patient
suggestive of AIN and should prompt a careful review of medication responds to 60 mg of IV furosemide bolus, he or she may be converted
exposure and consideration for kidney biopsy and/or treatment. to a 10 mg/h continuous infusion. Continuous infusions increase
Tuberculosis, chlamydia, ureaplasma, and mycoplasma are infectious the area under the curve of therapeutic efficacy, allowing for more
agents that may also cause sterile pyuria. consistent dosing. In one comparison study of heart failure patients
The urine sodium and FeNa are helpful in delineating prerenal there was a trend for increased dosage requirement with bolus dosing
physiology in AKI. Typically, a urine sodium under 20 mEq/L and compared with continuous therapy, and there was a trend for fewer
a FeNa under 1% are markers of a prerenal state and are usually complications with continuous infusion as well (Felker et al., 2011).
associated with a bland UA (Table 87.6). The FeNa is superior to Bumetanide and torsemide are alternative loop diuretic agents
measurement of urinary sodium alone because it does not vary by and have the advantage of increased potency and improved oral
urinary volume. Low sodium and FeNa may also be seen in shock, bioavailability compared with furosemide. Torsemide also exhibits
heart failure, HRS, contrast nephropathy, rhabdomyolysis, or acute a longer duration of action compared with furosemide. Comparison
GN. One caveat to urinary sodium analysis is that patients on diuretic of dosing of loop diuretics is shown in Table 87.7. Ethacrynic acid
therapy may have higher values despite volume depletion. An alterna- is occasionally used when patients have exhibited drug allergies to
tive measurement that may be undertaken is the fractional excretion the sulfonamide component of loop diuretics. However, ethacrynic
of urea (FeUrea) (see Table 87.6). FeUrea is typically less than 35% acid may be more ototoxic than the sulfonamide diuretics at high
in prerenal disease and 50% to 65% in ATN (Carvounis et al., 2002; doses, and it is difficult to administer intravenously because of the
Pépin et al., 2007). Renal reabsorption of urea relies on intact proximal relative insolubility. Patients who fail to respond to high doses of
tubular function and may not be accurate in the face of hyperglycemia loop diuretics may respond to the addition of a low dose of thiazide
or other causes of osmotic diuresis. diuretic, such as metolazone 2.5 to 5 mg/d. Distal sodium absorp-
Imaging is typically incorporated in the evaluation of AKI. As tion may be augmented, particularly in patients on chronic loop
mentioned earlier, renal ultrasound is instrumental in identifying diuretic therapy, and an impressive increase in diuresis may be
most cases of obstructive uropathy and can also identify chronic observed by combining loop and thiazide diuretic therapy. Close
changes that may be seen after prolonged kidney injury and CKD. monitoring of electrolytes is essential with such dual diuretic therapy
Total kidney size reduction and loss of cortical echogenicity correlate because hypokalemia and metabolic alkalosis are common side
with tubular atrophy and interstitial fibrosis and may guide interven- effects. If there is minimal response to high-dose loop diuretic therapy
tion. For example, a chronically obstructed kidney that is small and combined with a thiazide in an oliguric patient, then dialysis or
atrophied may not recover significant function after ureteral stenting ultrafiltration therapy should be considered.
or surgical intervention. CT imaging with low radiation dose is a Other agents such as low-dose dopamine and fenoldopam
highly sensitive test to identify or confirm stone disease and can be have not been shown to improve outcomes in AKI. One randomized
used in the absence of IV contrast in patients with AKI. CT has trial assigned 328 critically ill patients to low-dose dopamine (2 μg/
kg/min) versus placebo. There were no differences in renal recovery,
need for dialysis, hospital stay, or mortality between groups (Bellomo
TABLE 87.6 Formulas for the Fractional Excretion of et al., 2000). Even “renal” dose dopamine may precipitate cardiac
Sodium and Urea arrhythmias and should be used with caution. A large randomized
trial using fenoldopam, a selective dopamine receptor-1 agonist,
FeNa, % = UNa × SCr / SNa × UCr × 100 similarly did not show any improvement in AKI compared with
FeUrea, % = Urea × SCr / SBUN × UCr × 100 placebo (Bove et al., 2014). The study included 667 cardiac surgical
patients with early AKI who were randomized to fenoldopam versus
B, Blood; BUN, blood urea nitrogen; Cr, creatinine; Fe, fractional excretion; placebo. There were no differences in rates of dialysis or mortality,
Na, sodium; U, urine. and patients on fenoldopam had a higher rate of hypotension.

Downloaded for FK UMI Makassar (mahasiswafkumi09@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier on January 10, 2021.
For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Chapter 87 Renal Insufficiency and Ischemic Nephropathy 1927

TABLE 87.7 Comparison of Dosing, Bioavailability, and Half-Life of Various Loop Diuretics

DRUG BIOAVAILABILITY EQUIVALENT DOSING DURATION OF ACTIVITY DOSING INTERVAL

Furosemide oral 50% with variability 40 mg 6h Daily to q6h


Furosemide IV 100% 20 mg 6h Daily to q6h
Bumetanide oral 80%–90% 1 mg 6h Daily to q12h
Bumetanide IV 100% 1 mg 6h Daily to q12h
Torsemide oral 80%–90% 20 mg 8–12 h Daily to q12h
Torsemide IV 100% 20 mg 8–12 h Daily to q12h
Ethacrynic acid oral 100% 50 mg 6h Daily to q8h
Ethacrynic acid IV 100% 50 mg 6h Daily to q8h

IV, Intravenous.

Electrolyte abnormalities are common after AKI, and hyper- therapy, and hypotension often occurs during therapy. There may
kalemia is the most common and dangerous electrolyte abnormality be a greater risk for ischemic injury during dialysis treatment, and
in the AKI setting (Fordjour et al., 2014; Hoorn et al., 2013). If efforts should be made to avoid exacerbating hypotension and
serum potassium levels exceed 6 mEq/L, an electrocardiogram (ECG) hemodynamic instability. Slower, continuous dialysis modalities are
should be obtained and evaluated. Peaked T waves may be observed often used in the ICU setting, particularly in hypotensive patients
with broadening of the PR interval, which may lead to QRS broadening or in patients dependent on pharmacologic vasopressor support.
and the potential for maturation into a sine waveform. The goals
for treatment of hyperkalemia in the face of ECG changes include
stabilization of the cardiac conduction system, shifting potassium KEY POINTS: ACUTE KIDNEY INJURY
intracellularly, and eventual elimination of excess potassium from • AKI is defined by an acute rise in SCr and/or an acute
the body. Stabilizing the cardiac membrane may be achieved by decline in urine output. This abrupt decline in kidney
immediate administration of calcium salts. Shifting potassium into function occurs over hours to days and results in the
cells may be accomplished by insulin therapy (usually with glucose accumulation of byproducts of metabolism and the
to prevent hypoglycemia), sodium bicarbonate, or β2-adrenergic dysregulation of electrolyte homeostasis, acid/base, and
agonist therapy. A cationic binding resin such as sodium polystyrene volume status.
sulfonate (SPS) may be used to augment potassium excretion in the • A classic way to divide AKI is by “prerenal,” “intrinsic,”
gut. SPS appears to have modest efficacy (Hagan et al., 2016). There and “postrenal” or obstructive causes. The hallmark of
are reported cases of intestinal necrosis with SPS, with or without prerenal disease is its reversibility after treatment of the
the concomitant use of sorbitol (Goutorbe et al., 2011). Its use should underlying cause, with absence of structural damage to the
be avoided in patients with postoperative ileus, intestinal obstruction, kidney if treatment promptly.
or active colitis. • Prolonged prerenal physiology may lead to intrinsic
Two newer agents have been approved that augment the excre- kidney ischemia and injury. The greatest proportion of
tion of potassium and are the first agents available since SPS was hospital-acquired AKI is secondary to ATN.
approved for use in 1958. Patiromer is a nonabsorbed polymer • Nephrotoxic pharmacologic agents are a common source
that exchanges potassium for calcium in the gastrointestinal tract. of kidney injury through direct toxic effects or by causing
It has efficacy in dropping potassium levels over days by approximately inflammation (interstitial nephritis).
0.75 mEq/L in patients with CKD, with minimal adverse effects • Examination of the UA and urinary studies, including
including constipation and hypomagnesemia (Bushinsky et al., 2015; measurement of electrolytes and protein concentration, are
Weir et al., 2015). It is taken in powder form mixed with water and fundamental to the evaluation of the patient with AKI.
must be separated from other oral medications. However, because
of its delayed onset of action, patiromer is not indicated for the
treatment of severe acute hyperkalemia. A second agent, sodium CHRONIC KIDNEY DISEASE
zirconium cyclosilicate (ZSC), was approved for use in the United
States in 2018. It is also a nonabsorbed inorganic cation exchange CKD is defined as kidney impairment sustained beyond 3 months
agent that works in the gastrointestinal tract by exchanging potas- with a reduction in GFR, structural abnormalities in the kidneys,
sium for hydrogen and sodium. It also can lower potassium over and/or proteinuria (often defined specifically by albuminuria).
48 hours and is not approved for emergent treatment of severe As described earlier, estimations in GFR may be derived from the
hyperkalemia but holds promise as an agent with minimal adverse MDRD and CKD-EPI equations and should be used when kidney
effects (Hoy, 2018). function and SCr are at steady state. The 2012 KDIGO guidelines
Despite medical intervention, dialysis therapy may be required included albuminuria in the definition of CKD to emphasize the
for patients with severe, persistent AKI. The indications for initiation prognostic implications related to abnormal urinary protein excretion.
of dialysis therapy include volume overload, severe hyperkalemia, For example, a patient with diabetic nephropathy who has a GFR
severe metabolic acidosis, pericarditis, selected poisonings and drug of 60 mL/min/1.73 m2 and is excreting 3 g/day of albuminuria is
overdoses, and uremic symptomatology. There are disparate outcomes more likely to progress to ESRD than a patient with a GFR of 45 mL/
from studies examining the utility of starting dialysis earlier in the min/1.73 m2 related to prior kidney injury and no albuminuria.
course of AKI in the ICU setting. One study in patients with septic The interplay between albuminuria and GFR as outlined by the 2012
shock and advanced failure by RIFLE criteria (see Table 87.1) were KDIGO guidelines is shown in Fig. 87.1.
randomized to early renal replacement therapy within 12 hours The presence of CKD has implications in terms of greater risk
versus delaying more than 48 hours if there was no urgent indication for AKI, and recognizing that CKD is a risk factor for AKI should
for dialysis. Mortality rates were similar at 90 days between groups, lead to measures taken to prevent AKI in the hospital or office setting.
and dialysis was avoided altogether in 38% of the delayed group Precautions should be taken in CKD patients to avoid nephrotoxic
(Barbar et al., 2018). A theoretical concern exists that the dialysis drugs, avoid or limit iodinated contrast exposure, and to dose
treatment may have a detrimental impact on recovery of AKI. A drop medications appropriately with adjustment for level of kidney func-
in urine output is often seen during and after intermittent dialysis tion. Conversely, it is important to recognize that AKI often leads

Downloaded for FK UMI Makassar (mahasiswafkumi09@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier on January 10, 2021.
For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
1928 PART VIII Renal Physiology and Pathophysiology

CURRENT CHRONIC KIDNEY DISEASE (CKD) NOMENCLATURE USED BY KDIGO

CKD is defined as abnormalities of kidney structure or function, present for more than 3 months, with
implications for health, and CKD is classified based on cause, GFR category, and albuminuria category (CGA).
Prognosis of CKD by GFR and albuminuria category

Persistent albuminuria categories


description and range

Prognosis of CKD by GFR A1 A2 A3


and albuminuria categories:
KDIGO 2012 Normal to mildly Moderately Severely
increased increased increased
<30 mg/g 30–300 mg/g >300 mg/g
<3 mg/mmol 3–30 mg/mmol >30 mg/mmol

G1 Normal or high ≥90


GFR categories (mL/min/1.73 m2)

G2 Mildly decreased 60–89


description and range

Mildly to moderately
G3a 45–59
decreased
Moderately to
G3b 30–44
severely decreased

G4 Severely decreased 15–29

G5 Kidney failure <15

Green: low risk (if no other markers of kidney disease, no CKD); Yellow: moderately increased risk;
Orange: high risk; Red: very high risk.

Fig. 87.1. Chronic kidney disease (CKD) risk for progression based on Kidney Disease: Improving Global
Outcomes (KDIGO) 2013 classification of glomerular filtration rate (GFR) and albuminuria. (From KDIGO
Committee: KDIGO 2012 Clinical Practice Guideline for the Evaluation and Management of Chronic Kidney
Disease. Kidney Int Suppl 3[1]:2012.)

to CKD that will require monitoring after renal recovery (Lewington TABLE 87.8 Incidence of Reported Causes of
et al., 2013; Pannu, 2013). End-Stage Renal Disease
Deficits in nephron number may predispose to progressive kidney
disease and hypertension. Nephron number averages approximately DIAGNOSIS PERCENTAGE
600,000 ± 200,000 per normal kidney. Loss of kidney mass through
ablation or partial nephrectomy may trigger progressive glomerular Diabetes mellitus 38
injury in the remnant kidney. The injury is associated with hyperfiltra- Hypertension 25
tion, glomerular hypertrophy, and systemic hypertension (Brenner Glomerulonephritis 14
and Mackenzie, 1997). Proteinuria appears to correlate with degree Cystic kidney disease 5
of nephron loss and time from the loss (Novick et al., 1991). Reduced Urologic disease 2
birth weight or solitary kidney from birth may similarly lead to Other/missing/unknown 15
kidney injury over time because of low nephron number (Bhathena
et al., 1985; Hughson et al., 2003; Lopes and Port, 1995). In general,
monitoring of kidney function, albuminuria, and blood pressure
should be undertaken after loss of kidney mass. This includes living diagnosis. This is particularly common in the black community, in
kidney donors, although the typical natural history in kidney donors which the diagnosis of hypertensive kidney failure is increasingly
is an improvement in GFR and lack of proteinuria in the years after being recognized to correlate with genetic risk of CKD.
donation (Fehrman-Ekholm et al., 2011). The development of Accordingly, special mention should be made to the identifica-
albuminuria should lead to targeted therapy in patients with reduced tion of genetic risk for CKD in a subset of black patients, which
renal mass. The use of ACE inhibitors or ARBs appears to be protective relates to variants in apolipoprotein genetics. Polymorphisms
and can reduce proteinuria in a “remnant kidney” model (NDT 9: in the genes that encode apolipoprotein L1 (APOL1) are found
131, 1994), (Novick and Schreiber, 1995). in patients of sub-Saharan African ancestry, and two separate vari-
The causes of progressive CKD parallel the most common causes ants have been identified: G1 and G2. These gene variants appear
of ESRD. Table 87.8 lists the incident causes of ESRD from the US to convey protection against African sleeping sickness caused by
Renal Data System (USRDS, 2013). Diabetes mellitus and hyperten- Trypanosoma parasitic infection, whether inherited as a single or dual
sion account for the largest percentage of cases (63%) followed by variant (Vanhamme et al., 2003). Inheritance of two APOL1 gene
glomerular diseases (14%). A caveat to such reporting is that many variants of any combination (G1/G1, G1/G2, or G2/G2) appears to
patients go without kidney biopsy and lack a definitive cause of greatly increase the risk of CKD related to hypertensive kidney
ESRD. Genetic disease or other occult kidney diseases may com- disease, focal sclerosis, diabetic nephropathy, lupus nephritis, and
monly go undiagnosed or unrecognized, resulting in the labeling human immunodeficiency virus associated nephropathy (HIVAN) in
of “hypertensive” kidney disease in the absence of a definitive patients of African descent (Cohen et al., 2017; Larsen et al., 2013;

Downloaded for FK UMI Makassar (mahasiswafkumi09@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier on January 10, 2021.
For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Chapter 87 Renal Insufficiency and Ischemic Nephropathy 1929

Parsa et al., 2013). Two gene variants are found in approximately to guide treatment of membranous nephropathy because clearance
13% of the black population (Friedman et al., 2011) and explain to of the antibody often precedes a reduction in proteinuria and is
a large extent the increased risk of CKD and ESRD seen in blacks predictive of remission (Beck et al., 2011). Reemergence of antibodies
in the United States (Freedman et al., 2018). Although APOL1 is a may also predict disease recurrence (Ruggenenti et al., 2015). Other
circulating protein, it has also been localized to the podocyte, proximal antibodies have been identified that are present in a minority of
tubule, and vasculature of the kidney (Madhavan et al., 2011). The membranous cases, including one targeting thrombospondin type-1
idea that the integrity of the kidney may be affected by APOL1 gene domain-containing 7A (THSD7A), another podocyte transmembrane
variation is suggested by studies in kidney transplantation, which protein. THSD7A antibodies have been detected in 10% of patients
demonstrate reduced allograft survival when kidneys came from with membranous who lack antibodies to PLA2R (Ren et al., 2018).
deceased donors with high-risk gene variants (Freedman et al., 2016). Some cases of membranous nephropathy with antibodies against
The long-recognized risk of inferior transplant outcomes with kidneys THSD7A have been associated with malignancy, and clearance of
from black deceased donors also appears to be entirely explained by the antibody leading to remission of glomerular disease has been
the disproportionate risk related to the smaller percentage of donors described with the use of chemotherapy targeting the malignancy
with two APOL1 gene variants (Julian et al., 2017). directly (Hoxha et al., 2016).

Glomerulonephritis End-Stage Renal Disease Demographics


and Treatment Options
Glomerulonephritis is evolving in terms of accuracy of diagnosis
and identification of cause. The diagnosis of focal segmental glo- There continues to be a steady growth of ESRD patients on renal
merulosclerosis (FSGS) may represent a primary immune process replacement therapies (RRTs) throughout the world. In the United
or a secondary pattern of injury. Secondary forms of FSGS may States the incidence rate for ESRD in 2016 was 373 new cases per
develop for many reasons, including loss of renal mass such as after million people (US Renal Data System, 2018). The prevalence of
partial or total nephrectomy, genetic risk such as APOL1 gene variants ESRD patients has continued to increase since 2006, and the number
in blacks, a consequence of morbid obesity (Goumenos et al., 2009), of patients with ESRD in the United States in 2016 was approximately
or as a consequence of other prior drug or immune-mediated injury. 726,000. The number of kidney transplants performed annually in
In addition, primary FSGS may be familial rather than immune the United States has increased slightly and in 2016 was just more
mediated. Familial FSGS is suggested by a positive family history of than 20,000, with approximately one-third of transplants coming
disease and an onset of nephrotic syndrome at a very young age from living kidney donors. Rates of living donation have declined
(Sadowski et al., 2015). Identifying such secondary or genetic forms slightly since 2004 and remained relatively level in more recent
of FSGS is critical because these patients would not be expected to years. In 2016, 87% of patients with incident ESRD in the United
benefit from immunotherapy with medications such as corticosteroids States started hemodialysis, 10% started on peritoneal dialysis, and
or calcineurin inhibitors. Diffuse podocyte effacement on electron 3% received a preemptive kidney transplant (before any dialysis
microscopic biopsy in the absence of genetic association may therapy). By the end of 2016, 63% of prevalent ESRD patients were
alternatively point to a primary FSGS for which immunotherapy on hemodialysis, 7% were on peritoneal dialysis, and 30% had a
would potentially convey benefit (De Vriese et al., 2018). Such primary functioning kidney transplant. Although home hemodialysis is a
FSGS patients are more likely to be young adults, Caucasian, and modality that has demonstrated improved survival (Mailloux et al.,
have heavy proteinuria in the nephrotic range. 1996) and cost effectiveness (McGregor et al., 2000), only 2% of
Nephrotic syndrome is a clinical presentation that may be caused hemodialysis patients received home treatments in 2016.
by FSGS or other primary or secondary glomerular diseases. It is Poor long-term survival of ESRD patients is illustrated by compar-
characterized clinically by a urinary protein excretion of greater than ing the expected remaining lifetime on dialysis or transplant with
3.5 g/day, hypoalbuminemia, and peripheral edema. Primary diseases the general US population. The expected survival for dialysis patients
that may cause nephrotic syndrome include FSGS; minimal change was less than one-third of those age-matched in the general popula-
disease, which is especially common in children; membranous tion, whereas the expected survival in transplant patients was 69%
nephropathy; and certain types of membranoproliferative diseases. to 85% of that in the general population. The adjusted mortality
Secondary nephrotic syndrome may be seen in secondary FSGS related rate in patients on dialysis was 164 per 1000 patient-years, whereas
to infection such as HIV infection, lupus-associated membranous the rate in transplant recipients was 29 per 1000 patient-years.
nephropathy, hereditary diseases, and amyloid or light chain deposi- Importantly, however, survival is improving for patients on dialysis
tion caused by monoclonal proteins. and with transplantation. Between 2001 and 2016, adjusted mortality
Abnormal monoclonal proteins in the absence of defined rates decreased in dialysis patients by 29%. Such improved survival
myeloma or amyloidosis may be seen with variable nephrotic is contributing to the growing prevalence of ESRD patients. This
and nephritic presentations in the kidneys and has been termed improvement in survival was similar to that seen in non-dialysis
monoclonal gammopathy of renal significance (MGRS) (Leung et al., patients on Medicare with diabetes and cancer. In transplant patients,
2012). Accordingly, testing for monoclonal proteins as a potential the adjusted mortality rates between 2001 and 2016 decreased by
cause of glomerular disease is essential, with risk for MGRS increasing 40%. The transplant population is aging. In 2001, 8% of transplant
with age. Serum and urine electrophoresis along with measurements recipients were over the age of 65, and in 2016 the rate increased
of free light chains kappa and lambda may detect low level mono- to 24% (USRDS, 2018).
clonal proteins that may be involved in renal glomerular or tubular Outcome comparisons suggest that kidney transplantation is
pathology. Treatment with agents used to treat myeloma may be still the best overall treatment for ESRD patients, despite advances
indicated even in the absence of other organ disease if renal pathology in dialysis care and modalities. The time on dialysis (vintage) has
is present (Sethi and Rajkumar, 2013). an impact on transplant outcomes, with inferior outcomes seen with
Membranous nephropathy is one of the most common causes longer dialysis vintage (Meier-Kriesche et al., 2000). This finding
of nephrotic syndrome in adults. The diagnosis and treatment of likely relates in part to progressive vascular disease on dialysis therapy
membranous nephropathy have improved because of the discovery but also reflects socioeconomic status and timeliness of listing (Schold
of a target antibody against a podocyte protein that is positive et al., 2010). Patients who progress to stage IV CKD with an estimated
in 70% to 80% of idiopathic membranous disease cases. The or measured GFR of 20 mL/min/1.73 m2 or less are eligible for active
antibody targets the M-type phospholipase A2 receptor (PLA2R), transplant listing in the United States and should be counseled and
a transmembrane receptor that is highly expressed in glomerular referred for transplant evaluation. Patients at advanced age or with
podocytes (Beck et al., 2009). This antibody may be identified in severe medical comorbidities or problems with medical adherence
the glomerulus on kidney biopsy by immunofluorescence or immu- may be excluded from transplant listing. For those approved, preemp-
nohistochemistry staining and may be detected in the circulation tive transplantation, before initiation of dialysis therapy, should
by ELISA or other methods. Monitoring of PLA2R Ab titers is useful be the goal. Because of long waiting times for deceased donor

Downloaded for FK UMI Makassar (mahasiswafkumi09@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier on January 10, 2021.
For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
1930 PART VIII Renal Physiology and Pathophysiology

transplantation, preemptive transplant is most likely to be achieved 110.0


with kidney donation from a living donor. 100.0

Available blood flow (%)


90.0

Atherosclerotic Renal Artery Stenosis 80.0


70.0
Atherosclerotic renal artery stenosis, a recognized cause of severe, 60.0 Systolic blood flow (%)
secondary hypertension and impaired renal function, is present in 50.0 Diastolic blood flow (%)
approximately 7% of individuals older than 65 years (Hansen et al., 40.0
2002) and in 40% to 50% of patients with atherosclerotic disease 30.0
in other vascular beds (Miralles et al., 1998). Clinically relevant 20.0
complications, specifically resistant hypertension and/or renal atrophy 10.0
with progressive decline in renal function, develop in 15% to 20% 0.0
of cases of atherosclerotic renal artery stenosis (Caps et al., 1998; 0 10 20 30 40 50 60 70 80 90 100
Chabova et al., 2000). In addition, atherosclerotic renal artery stenosis % Area stenosis
appears to be an independent risk factor for adverse cardiovascular
events and increased cardiovascular mortality. In 3 recent studies, Fig. 87.2. Relationship of blood flow to increasing arterial stenosis.
patients with atherosclerotic renal artery stenosis had between a
two- and fivefold increases in mortality compared with healthy
age-matched normotensive control patients (Conlon et al., 2001; and fluid viscosity (η) and is inversely proportional to the second
Edwards et al., 2005; Johansson et al., 1999). Moreover, the increased power of the radius of the tube.
risk profile appears to remain despite anatomic correction of ath-
erosclerotic renal artery stenosis. Pouiseuille’s law = ∆P = Q8Lη r 2
Atherosclerotic renal artery stenosis accounts for 90% of the
lesions that impede blood flow within the renal artery. Fibromuscu- Viewed simplistically, this relationship shows that as the radius (or
lar dysplasias, vasculitides, congenital bands, and intrinsic and extrinsic diameter) of a tube decreases, the difference in pressure increases
causes make up the remaining 10% of causes. Atherosclerosis typically exponentially. Therefore, as the diameter of a blood vessel decreases,
involves the ostium and proximal one-third of the renal artery and can the decline in pressure, ΔP, increases.
also involve the adjacent aorta. Involvement of segmental renal arteries The second important concept relates to the relationship between
and higher-order branches is uncommon. Because atherosclerosis luminal diameter and cross-sectional area. The cross-sectional area
is a systemic condition, atherosclerotic renal artery stenosis most of a tube, which is equal to that of a circle, is expressed as A=πr2.
often occurs in patients with risk factors for atherosclerosis, such as This can be further expanded to consider diameter or A=π (d/2)2 or
hypertension, hyperlipidemia, diabetes mellitus, and tobacco use. πd2/4. Again showing that as the diameter of a vessel decreases, the
Early studies determined the prevalence of renal artery disease in luminal area decreases by the square of the diameter reduction. Early
autopsy series. In patients older than 60 years and with a history of experiments have conclusively shown that clinically significant vascular
hypertension, moderate to severe renal artery stenosis was found in stenosis, specifically those that cause a pressure drop, occur when
approximately 50% of patients (Holley et al., 1964; Schwartz et al., the luminal area is reduced by more than 70%, also expressed as
1964). Because atherosclerosis-related complications are a leading “70% stenosis” (May et al., 1963; Fig. 87.2). This corresponds to a
cause of death, autopsy studies may overestimate the prevalence of diameter reduction of approximately 50% because such a diameter
atherosclerotic renal artery disease but nonetheless suggest a direct reduction will result in a reduction in cross-sectional area to 25%
relationship to increasing patient age and severity of hypertension. of the original area.
The clinical prevalence of atherosclerotic renal artery stenosis, Given that a constant amount of fluid (blood) circulates through
which is frequently identified in patients with cerebrovascular, coro- the vascular system, another basic assumption is that blood flow
nary, and peripheral arterial disease depends on the radiographic (volume of blood per time) is maintained in all parts of the circulatory
definition (percent of renal artery luminal narrowing), imaging system. Extending from the Bernoulli equation, as the diameter of
technique, and the patient population being studied. In a general tube decreases, the velocity of the moving fluid increases to maintain
population of patients older than 65 years of age the prevalence was the conservation of energy principle of flowing fluids. Importantly,
noted to be 6.8%, when assessed with duplex Doppler ultrasonography these concepts are based on laminar flow of frictionless fluids through
(Hansen et al., 2002). In a systematic review of studies using more rigid tubes, markedly different from the human circulatory system.
sensitive imaging techniques and defining atherosclerotic renal artery Flow is maintained in human arteries by increasing velocity until
stenosis as at least 50% luminal narrowing, the prevalence ranged a critical level of stenosis or approximately a 70% decrease in area.
between 10.5% in consecutive patients undergoing coronary angi- Beyond 70%, flow begins to decrease rapidly, and beyond a 90%
ography to 54.1% in patients with congestive heart failure (de Mast stenosis flow and velocity are reduced, compatible with “trickle-flow”
et al., 2009). In patients with underlying atherosclerotic conditions, (Fig. 87.3).
atherosclerotic renal artery stenosis can be anticipated in approxi- Based upon this relationship, flow decreases markedly at critical
mately 10% to 20% of cases. With these sensitive imaging techniques levels of stenosis (>70%). Under normal systolic blood pressures
the prevalence of bilateral atherosclerotic renal artery stenosis is (120 mm Hg) there is an approximately 40- to 60-mm Hg pressure
4.2%, in contrast to 0.8% when identified by duplex ultrasonography drop to the level of the afferent arterioles within the kidney (Fig. 87.4).
(de Mast et al., 2009; Hansen et al., 2002). De Bruyne et al. (2006) have shown that as the degree of stenosis
increases to approximately 50%, the pressure in the renal artery
Pathophysiology of Hypertension in Atherosclerotic Renal distal to stenosis is 60 to 70 mm Hg. It is likely therefore that at
Artery Stenosis this lower perfusion pressure, the pressure and flow, at the afferent
arteriole reaches precipitously low levels, thus initiating the patho-
Basic Concepts in Arterial Physiology. Blood flows throughout physiologic processes.
the circulatory system primarily because of differences in total fluid The pathophysiologic basis for hypertension resulting from
energy but from a functional standpoint, blood flow can be attributed impaired renal blood flow was first elegantly described more than
to differences in pressure gradients. A pressure gradient is maintained 75 years ago (Goldblatt et al., 1941). These initial studies stimulated
by cardiac contractions and elasticity of the arterial system. The basic numerous experiments that outlined the hormonal basis for the
concept that describes the relationship between blood pressure (and/ kidney’s role in blood pressure control. Adding to these initial studies,
or flow) and blood vessel diameter follows Pouiseuille’s law, which it is now well outlined that a reduction in renal perfusion leads to
states that the pressure difference between two points along a tube a decrease in glomerular filtration. Consequently, the reduction in
is directly proportional to the flow velocity (Q), length of tube (L), distal tubular sodium chloride (NaCl) delivery to the macula densa

Downloaded for FK UMI Makassar (mahasiswafkumi09@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier on January 10, 2021.
For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Chapter 87 Renal Insufficiency and Ischemic Nephropathy 1931

600 perfusion pressure, which, in turn, promotes a natriuresis and volume


loss. Therefore maintenance of hypertension in unilateral renal
artery stenosis and normal contralateral kidney is driven primarily
500 by renin and angiotensin. In contrast, in the setting of bilateral
Flow (mL/min) – velocity (cm/sec)

renal artery stenosis or renal artery stenosis affecting a solitary


Velocity
kidney, sodium and volume retention are not modified to the
400
extent of having a normal contralateral kidney. Renin and angio-
tensin levels do not remain elevated, and hypertension remains
Flow sodium and volume dependent.
300
Given the transient elevation in renin and angiotensin, several
other pressor pathways participate in the development of hypertension.
These pathways include sympatho-adrenergic activation, oxidative
200
stress pathways, and impaired vasodilatory responses within the
kidney and systemic microcirculation (Lerman et al., 2001).
100
Pathophysiology of Ischemic Nephropathy in Atherosclerotic
100 90 80 70 60 50 40 30 20 10 0 % Decrease in area
Renal Artery Stenosis
80 70 60 50 40 30 20 10 0 % Decrease in diameter A reduction in renal perfusion resulting from atherosclerotic renal
artery disease, which in turn leads to renal parenchymal loss and
Fig. 87.3. Correlation of percent diameter reduction with increases in blood declining renal function, describes a condition known as ischemic
flow velocity (cm/sec) and reduction in volume flow (mL/min) in arteries. Note nephropathy. Several interrelated pathophysiologic mechanisms are
that a high-grade (>95%) diameter-reducing stenosis causes volume flow to likely responsible for the renal structural and functional changes
decrease toward zero, whereas the velocity within the stenosis may be minimally that occur in atherosclerotic renal artery disease. The impairment in
elevated. microvascular blood flow, in combination with conditions that
contribute to the atherosclerotic milieu, such as hypercholesterolemia,
diabetes mellitus, and hypertension, lead to vascular dysfunction,
120 which is mediated by endothelial injury, reduced bioavailability of
BP Interlobular nitric oxide, and increased vascular tone resulting from the predomi-
artery nance of vasoconstrictors, primarily angiotensin II. Concomitant
100
cellular activation leads to migration of mononuclear cells and release
Pressure (mm Hg)

Arcuate inflammatory mediators including nuclear factor kappa-B (NF-κB).


80
artery Upregulated tubular and glomerular expression of profibrotic factors,
Afferent including transforming growth factor β (TGF-β), tissue inhibitor of
60 metalloproteinase-1, and plasminogen activator inhibitor-1 leads
arteriole Peritubular
Glomerular capillaries Renal to structural changes of tubulointerstitial fibrosis, vascular changes
40 and/or glomerulosclerosis (Chade et al., 2003; Lerman et al., 2009).
capillaries venules
Changes in intrarenal blood flow have been identified using blood
Efferent oxygen level–dependent (BOLD) magnetic resonance imaging (MRI)
20
arteriole
analysis. Gloviczki et al. (2013) found that in cases of severe renal
0 artery stenosis in which tissue hypoxia may occur, there is preferential
redistribution of blood flow away from the renal cortex and toward the
Fig. 87.4. Hydraulic pressure profile in the renal vasculature based on a variety medulla. At the same time, there are changes in metabolic processes
of micropuncture studies in superficial nephrons of the rat and squirrel monkey to preserve oxygen (Eirin et al., 2013).
as well as values obtained by micropuncture of juxtamedullary nephrons in Studies sampling renal vein blood from patients with atheroscle-
the rat. For these latter studies, the arcuate artery was perfused with whole rotic renal artery stenosis compared with those with essential
blood at normal arterial pressures, and hydraulic pressures were measured hypertension found multiple markers reflecting active inflammation,
at downstream sites, including the interlobular artery, the proximal and distal suggesting kidney injury and reduced function. Endothelial progenitor
portions of the afferent arteriole, the glomerular capillaries, the proximal and cells expressing the markers CD34 and KDR were sequestered in
late segments of the efferent arteriole, the peritubular capillaries, and the kidneys with atherosclerotic renal artery stenosis, whereas levels of
renal vein. E-selectin, vascular cell adhesion molecule-1, and several inflammatory
molecules were increased (Eirin et al., 2013).
In summary, atherosclerotic renal artery stenosis, in combination
cells of the juxtaglomerular apparatus initiates an epithelial response with other atherosclerotic risk factors, results in progressive loss of
that consists of activation of multiple mitogen-activated protein renal function through a complex interplay between inflammatory
kinases and stimulation of cyclooxygenase-2 activity and induction and immune responses, angiotensin II, oxidative stress, vascular
of cyclooxygenase-2 expression. This is subsequently followed by rarefaction, and fibrosis. These molecular and cellular level processes
the appearance in the juxtaglomerular interstitium of prostaglandin ultimately lead to renal atrophy and diminished renal function.
E2 and the suppression of adenosine. Prostaglandin E2 activates
receptors on granular cells which then results in adenylate cyclase Diagnostic Evaluation of Atherosclerotic Renal
activation and phosphokinase A-mediated renin secretory and Artery Stenosis
transcriptional activation (Schnermann et al., 2003).
Renin, in turn, cleaves hepatically-produced angiotensinogen to Clinical Features. The primary goal when undertaking diagnostic
produce angiotensin I, which is then converted by the angiotensin evaluation for suspected atherosclerotic renal artery stenosis is
converting enzyme in the pulmonary vasculature to angiotensin II. to identify patients who, if found to have a lesion, are most likely
In addition to causing hypertension via its potent vasoconstrictive to benefit from treatment. Given that most patients with athero-
effects, angiotensin II stimulates adrenal synthesis of aldosterone, sclerotic renal artery stenosis are asymptomatic, the decision to pursue
which further results in sodium and fluid retention. diagnostic assessment, particularly with invasive studies, should be
In the classic 2-kidney, 1-clip model, increased renin release from driven by the probability that renovascular disease is a significant
the affected kidney leads to hypertension and sodium retention. contributor to the patient’s condition and will result in potential
However, the contralateral (unaffected) kidney senses increased clinical consequences. If blood pressure and renal function are

Downloaded for FK UMI Makassar (mahasiswafkumi09@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier on January 10, 2021.
For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
1932 PART VIII Renal Physiology and Pathophysiology

satisfactorily controlled with medical therapy, further diagnostic ultrasonographic examination for renal artery stenosis should be
evaluation may not lead to treatment gains. done by an experienced technologist, and patients should undergo
In general, the clinical criteria that should prompt evaluation are an overnight fast to minimize bowel gas. Imaging is performed from
(1) early onset of hypertension (age < 30 years), (2) accelerated anterior and flank (lateral decubitus) approaches, with attention
hypertension, (3) severe hypertension, or (4) hypertension resistant directed to the perirenal aorta, looking for aneurysmal change,
to optimal medical therapy, (5) asymmetric kidney size with more stenosis, or occlusion. The lateral decubitus position may aid in the
than 1.5 cm difference in craniocaudal length, (6) unexplained loss obese patients and/or in the setting of excessive bowel gas. Peak
of kidney function, (7) rapid or recurrent decline in GFR in association systolic velocity in the aorta is recorded. Subsequently each renal
with systemic blood pressure reduction, and (8) decline in GFR (SCr artery is imaged, and velocities and waveforms are recorded from
at least 30% from pretreatment level) after initiation of ACE inhibitors the proximal, mid, and distal segments of the renal arteries. Duplex
or ARBs. In addition, although most likely unrelated to atherosclerosis, ultrasonography with color-flow and Doppler imaging is used to
all children with unexplained hypertension should be screened for determine the patency and phasicity of the renal veins. Velocity
renal artery stenosis. measurements within the superior, mid, and lower poles of the
Other clinical features that are often present in patients with ath- kidneys are used to calculate resistive indices and acceleration times.
erosclerotic renal artery stenosis are typical for patients with systemic Last, the duplex ultrasonographic study determines renal parenchymal
atherosclerotic disease. These include advanced age; atherosclerotic anatomic details including kidney size, parenchymal thickness and
risk factors, including hypertension, CKD, hyperlipidemia, diabetes echogenicity, and potentially unanticipated findings such as masses,
mellitus, tobacco exposure; and history of atherosclerotic disease in calculi, and/or hydronephrosis.
the coronary, cerebral, and/or the peripheral arterial circulation. On The most commonly measured duplex ultrasonographic parameters
physical examination these patients often have severe hypertension include peak systolic velocity (PSV), end diastolic velocity (EDV),
and may also have fewer common signs, such as diminished lower and adjacent aortic peak systolic velocity. The ratio of velocity in
extremity pulses, peripheral edema, and abdominal bruits. the main renal artery to the aorta (renal aortic ratio, RAR) and
To generate uniformity, the American Heart Association proposed resistive index within the renal parenchyma (RI = [(PSV−EDV)/PSV])
the following classification scheme in patients with renal artery disease can be derived from these measurements. In addition, arterial blood
(Rocha-Singh et al., 2008): flow characteristics such as tardus/parvus, acceleration time, and
acceleration index can be determined within the main, accessory,
Grade I: Renal artery stenosis present, but no clinical manifestations and intraparenchymal renal arteries.
(normotensive and normal renal function) The accuracy of duplex ultrasonography for the diagnosis of renal
Grade II: Renal artery stenosis present, but patients have medically artery stenosis varies according to predetermined velocity thresholds,
controlled hypertension and normal renal function which in turn, depend on individual vascular laboratory experience.
Grade III: Renal artery stenosis present and patients have evidence AbuRahma et al. (2012) reviewed their institution’s large experience
of abnormal renal function, medically refractory hypertension, and found that a PSV of 285 cm/s or a RAR of 3.7 alone had the
or evidence of volume overload best overall accuracy in diagnosing at least 60% stenosis when
compared with angiography. Using these thresholds, these investigators
Laboratory Features were able to achieve more than 80% accuracy. Lower velocity
thresholds, although more sensitive, were less accurate. These findings
Apart from decreased GFR, patients with atherosclerotic renal artery reinforce the notion that clinicians should recognize the expertise
stenosis typically do not have notable laboratory abnormalities. The in their individual vascular laboratories and use the data to screen
urine analysis is likely to be unremarkable. On routine chemistry patients who require more invasive examination.
panels, patients may have low or low normal serum potassium because Computed tomography (CT) and magnetic resonance imaging (MRI)
of aldosterone secretion. have sensitivity of more than 90% in detecting atherosclerotic renal
Ritchie et al. (2014) retrospectively reviewed 467 patients with at artery stenosis, and MRI has a specificity of 91% (Eklof et al., 2006).
least 50% atherosclerotic renal artery stenosis and noted that nearly CT and MRI provide excellent anatomic detail; however, the blood
half of their patient group was asymptomatic. One or more “high-risk flow changes that result from the stenosis cannot be determined. The
presentations,” defined as flash pulmonary edema, refractory hyperten- functional consequences of these lesions may be indirectly determined
sion (>140 mm Hg systolic and/or >90 mm Hg diastolic) despite use by the findings of parenchymal atrophy. Another drawback of CT is
of 3 or more different classes of antihypertensive agents, or rapidly the cost and need for intravenously administered radiographic contrast
declining kidney function (SCr level more than 1.2-fold or 1.14 mg/ agents. Because many patients with suspected atherosclerotic renal
dL greater than baseline within the previous 6 months) was present in artery disease may have co-existent renal insufficiency, administration
51% of the patients, and 30% to 40% had a history of angina, previous of iodinated contrast agents raises concerns of contrast nephropathy.
myocardial infarction, cerebrovascular accident/transient ischemic attack, Patients with estimated glomerular filtration rate (GFR) below 50 mL/
peripheral arterial disease, or diabetes (Ritchie et al., 2014) min should receive IV hydration before contrast administration and
These data underscore a guiding principle for diagnostic evalu- those with GFR below 30 mL/min should not receive iodinated contrast
ation in atherosclerotic renal artery stenosis. Patients who are as part of a diagnostic procedure (contrast should be reserved for use
asymptomatic, have normal renal function, and have satisfactory during a therapeutic procedure). Administration of IV gadolinium
blood pressure control likely do not need evaluation. Alternatively, for magnetic resonance angiography (MRA) in patients with GFR
patients who fail to improve or who decline while on medical below 30 mL/min should be avoided because it is associated with
therapy may benefit from additional diagnostic testing. the development of nephrogenic systemic fibrosis.
Renal arteriography, the “gold standard” for the diagnosis of
Radiographic Assessment of Atherosclerotic Renal atherosclerotic renal artery stenosis, can also provide detailed imaging
Artery Stenosis of the aorta, segmental renal arteries, and intrarenal vasculature.
Digital subtraction improves resolution and allows a reduction in
Radiographic evaluation of suspected atherosclerotic renal artery the amount of contrast material. Angiography is an invasive study
stenosis should begin with noninvasive imaging studies. Given the with risks of bleeding from arterial puncture, arterial dissection,
wide availability, low cost, ease to the patient, and diagnostic accuracy, arterial spasm, and thromboembolism. Additionally iodinated contrast
duplex ultrasonography should be considered the screening test material cannot be administered in patients with contrast allergy or
of choice. Duplex ultrasonography combines anatomic information significant renal dysfunction, and use of carbon dioxide may reduce
(B- or brightness-mode and color Doppler imaging) with acquisition the amount of contrast.
of blood flow. Two additional diagnostic studies that warrant discussion are
The primary diagnostic advantage of duplex ultrasonography is radionuclide renography and plasma renin determination. With
the ability to accurately determine blood flow velocity. A duplex radionuclide or nuclear renography, a radiopharmaceutical agent,

Downloaded for FK UMI Makassar (mahasiswafkumi09@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier on January 10, 2021.
For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Chapter 87 Renal Insufficiency and Ischemic Nephropathy 1933

either diethylenetriaminepentaacetic acid (DTPA) or mercaptoacet- between treatment groups, and did not use current medical therapy,
yltriglycine (MAG 3), is administered intravenously, and images are thus making the results difficult to apply to current practice.
reviewed to determine a renal functional difference, delay in time When compared with PTRA alone, percutaneous transluminal
to peak uptake of the radioisotope, or delayed excretion of the isotope. renal artery angioplasty with stenting (PTRS) was found to have
Additional imaging after administration of captopril, an ACE inhibitor, superior technical success rates, increased patency rates, and decreased
may show decreased function (GFR), suggesting the influence of restenosis rates, leading to widespread adoption of PTRS as the
angiotensin II maintaining glomerular filtration. Radionuclide standard endovascular management for atherosclerotic renal artery
renography has largely fallen out of favor because of its unreliability. stenosis during the 1990s and early 2000s (Liang et al., 2013; van
In addition to diagnostic limitations in patients with diminished de Ven et al., 1999). A comparison of PTRS and medical manage-
renal function (SCr > 2.0 mg/dL) or bilateral arterial disease, these ment for atherosclerotic renal artery stenosis has been the subject of
imaging studies have poor sensitivity and specificity (Vasbinder et al., several randomized controlled trials. In three of these trials, which
2001). Currently the primary role of renography in evaluating patients were presented or published in 2009, nearly 1000 patients with
with hypertension may be to determine differential function before atherosclerotic renal artery stenosis were enrolled and followed for
recommending nephrectomy. 2 to 4 years (Bax et al., 2009; Kumbhani et al., 2011; Wheatley et al.,
Plasma renin activity or determination of renin level from the 2009). Aside from a slight reduction in number of antihypertensive
renal vein has been suggested to be highly diagnostic for the presence medications, no significant improvement in systolic or diastolic blood
of renal artery stenosis, but these studies are limited by their invasive- pressure, renal function decline, or adverse cardiovascular events
ness and stringent testing conditions. The patient’s volume status was seen with PTRS and medical management in comparison with
must be optimized, concurrent medications must be addressed, and medical management alone. Despite the randomized prospective
blood pressure must be tightly controlled. Last, and as mentioned design, these trials have faced several criticisms, including discrepan-
previously, renin may be elevated early in the course of atherosclerotic cies in atherosclerotic renal artery stenosis diagnosis, flaws in the
renal artery stenosis and likely declines over time as other pathophysi- intention-to-treat analysis, and inclusion of patients with wide-ranging
ologic processes maintain the hypertensive state. degrees of stenosis.
The NIH sponsored Cardiovascular Outcomes in Renal Atheroscle-
Therapeutic Options for Atherosclerotic Renal rotic Lesions, or CORAL, trial attempted to correct these limitations
Artery Stenosis and definitively answer whether revascularization of hemodynamically
significant atherosclerotic renal artery stenosis in hypertensive patients
Medical Management. Medical management makes up the founda- prevents adverse cardiovascular and renal events when added to
tion of atherosclerotic renal artery stenosis treatment. The general optimal medical therapy (Cooper et al., 2014). The CORAL study
objectives of therapy are control of blood pressure, preservation of enrolled 947 patients with atherosclerotic renal artery stenosis
renal function, and prevention of atherosclerosis-related complica- and hypertension between 2005 and 2010 and randomly assigned
tions. Antihypertensive treatment with ACE inhibitors or ARBs are these patients to medical therapy plus renal artery stenting or
the first-line approach for blood pressure control and for their medical therapy alone. After a median follow-up period of 43
beneficial effect on cardiovascular risk reduction (Heart Outcomes months, the rate of the primary composite end point, made up of
Prevention Evaluation Study Investigators, 2000). Use of these agents cardiovascular and renal sequelae, was not significantly different
may initiate concerns of renal insufficiency from loss of glomerular between participants who underwent stent placement in addition
filtration as a result of blockade of the AII-induced efferent arteriolar to medical therapy versus those who received medical therapy alone
constriction. However, significant renal function decline is uncommon (Cooper et al., 2014). Although the limitations of these studies
in most series and, when it occurs, is almost diagnostic of renal continue to be debated, the conclusions reached by the CORAL
artery stenosis. If additional antihypertensive agents are needed, study reliably show the lack of benefit of renal artery stenting in
beta-blockers, diuretics, and calcium-channel blockers can be added. most patients with atherosclerotic renal artery stenosis.
Given the presence of systemic atherosclerosis, many patients Complications of PTRS, although generally less frequent than
with atherosclerotic renal artery stenosis also have pre-existing those seen with open surgical revascularization, can still account
cardiovascular risk factors, including essential hypertension, diabetes for significant morbidity. Periprocedural mortality rate is approxi-
mellitus, dyslipidemia, and smoking. Established additional related mately 0.7%, and renal artery complications range from 1% to 10%
therapies include glycemic control, statins, antiplatelet agents, and (Kumbhani et al., 2011).
lifestyle modifications, including tobacco cessation. In contrast to asymptomatic patients who are found to have
Procedural Management. Whether all patients with atherosclerotic high-grade atherosclerotic renal artery stenosis, a subgroup of patients
renal artery stenosis should undergo renal revascularization in addition who appear to benefit from renal artery revascularization are those
to medical therapy remains a controversial practice, although recent with flash pulmonary edema. As stated earlier, randomized prospective
trials do not support this procedure. Renal revascularization, with trials included mostly asymptomatic patients. Ritchie et al. (2014)
open surgical renal artery bypass and percutaneous transluminal evaluated outcomes between medical management alone compared
angioplasty with or without stent placement, has been the subject with medical management plus percutaneous renal artery revascu-
of several randomized prospective trials. Numerous single-center larization with stent placement in patients with high-risk clinical
retrospective studies have reported on various techniques of open presentation, defined as flash pulmonary edema, rapidly declining
surgical revascularization and have shown durable long-term patency renal function, or refractory hypertension. In this large analysis, flash
rates (Cambria et al., 1996; Cherr et al., 2002; Darling et al., 1999; pulmonary edema appeared to be an adverse prognostic marker
Fergany et al., 1995; Hansen et al., 2000; Marone et al., 2004; Paty with increased risk of death and cardiovascular events in medically
et al., 2001). Notwithstanding these results, open surgical renal artery treated patients, whereas the combination with renal artery revas-
repair is associated with considerable perioperative mortality (5%) cularization reduced the risk of death (Ritchie et al., 2014).
and morbidity (20%) rates and therefore is generally reserved for
young patients and those with hemodynamically significant athero-
sclerotic renal artery stenosis and significant juxtarenal or suprarenal Nonatherosclerotic Renal Artery Diseases
aortic disease (Edwards et al., 2009). Fibromuscular Dysplasia
Renal revascularization with percutaneous transluminal renal
artery angioplasty (PTRA) has been compared with medical therapy Fibromuscular dysplasia (FMD) is a nonatherosclerotic, noninflam-
in three randomized controlled trials, none of which showed improved matory vascular disease that most often affects small to medium
renal function, hypertension management, or survival with PTRA arteries. Unlike atherosclerosis, FMD mostly involves the mid- to
(Plouin et al., 1998; van Jaarsveld et al., 2000; Webster et al., 1998). distal renal artery and can affect segmental renal artery branches.
These trials enrolled small numbers of patients, excluded patients FMD has a female predominance and typically occurs in patients
with severe renal functional impairment, had extensive crossover between 20 and 60 years of age but can occur in older and pediatric

Downloaded for FK UMI Makassar (mahasiswafkumi09@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier on January 10, 2021.
For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
1934 PART VIII Renal Physiology and Pathophysiology

patient populations. Asymptomatic patients are most often diagnosed gradients and even provide virtual histology of the arterial lumen
on imaging studies. Symptomatic patients can experience sequelae (Prasad et al., 2009). CT and MRA are able to suggest the diagnosis
of arterial disease such as stenosis, occlusion, arterial dissection, or of FMD in main renal vessels but are limited by poor visualization
aneurysm. Although FMD can occur in any arterial bed in the body, along branch renal vessels.
involvement of the renal artery is most common. Management of Fibromuscular Dysplasia. The primary treatment
Epidemiology of Renal Artery Fibromuscular Dysplasia. Because goals in patients with renal artery FMD are control of hypertension
many patients with FMD remain asymptomatic, the true prevalence and prevention of secondary complications of hypertension. Because
of FMD is difficult to assess. The overall prevalence can be estimated ischemic nephropathy is not a sequela of FMD, preservation of renal
from the evaluation of renal donors, a suitable representation of function is not a consideration in the treatment of FMD. The primary
healthy adult population. The prevalence of FMD in renal donors factors in considering which patients with FMD merit treatment are
ranges between 2.6% to 6.6% with the variance most likely resulting patient age, temporal relationship between onset of hypertension
from differences in imaging technique and patient populations (Cragg and diagnosis of FMD, adequacy of blood pressure control with
et al., 1989; McKenzie et al., 2013; Neymark et al., 2000). Bilateral anti-hypertensive agents, and duration of hypertension.
renal artery involvement is seen in approximately one-third of FMD Asymptomatic, normotensive patients with renal artery FMD
cases (McKenzie et al., 2013; Olin et al., 2011). FMD has a marked should be followed for the development of hypertension. Young,
female preponderance: nearly 90% of cases occur in women. Several otherwise healthy patients in whom FMD is diagnosed proximate
causative factors, including hormonal and environmental causes, to the onset of hypertension should be referred for PTRA, which
have been proposed for the development of FMD; however, the provides immediate and long-term blood pressure control in nearly
prevailing thought is that FMD is idiopathic. 75% of patients (Davies et al., 2008; Trinquart et al., 2010). Patients
Histopathology of Fibromuscular Dysplasia. FMD is classified in whom hypertension has been present for many years should be
according to the affected layer of arterial wall; specifically, the intima, continued on antihypertensive medications as long as blood pressure
media, and adventitia. FMD involving the media occurs most control is satisfactory. Dopper ultrasound–based surveillance of kidney
commonly and can be further subdivided into medial fibroplasia, length and cortical thickness should be done once or twice per year.
perimedial fibroplasia, and medial hyperplasia. The latter two condi- If blood pressure control becomes difficult, medication side effects
tions are very uncommon, whereas medial fibroplasia accounts become intolerable, or renal size or function decrease, PTRA should
for 90% of all types of FMD. Medial fibroplasia is characterized be performed (Olin, 2011). Balloon angioplasty alone is a very effective
by multiple diaphragm-like thickened areas of media that result in treatment for renal artery FMD, thereby making stent placement
segments of arterial stenosis and poststenotic dilatation, typically unnecessary. Surgical revascularization is reserved for cases not
seen as a “string of beads” appearance on CT angiography (Fig. 87.5). amenable to percutaneous approaches such as those with aneurysms
Accounting for the remaining 10% of cases of FMD, intimal or FMD involving distal extrarenal branches. Indications for stent
fibroplasia is due to collagen deposition within the intima with placement in FMD are inability to eliminate a pressure gradient with
resultant fragmentation of the intimal layer. On angiography, intimal angioplasty alone and intimal dissection.
fibroplasia often appears as a concentric stenosis and, occasionally, Treatment success is fairly similar between PTRA and surgery.
as a long, diffuse narrowing. Approximately 50% of patients have cure of hypertension, but the
Clinical Presentation. As mentioned previously, the diagnosis of complications are markedly fewer after PTRA. Major complication
renal artery FMD is often made in asymptomatic patients undergoing rates after PTRA are approximately 12%, in contrast to 17% after
abdominal imaging for other reasons. The most common symptomatic surgery (Trinquart, 2010).
presentation is a middle-age woman with new-onset or difficult- Restenosis can occur after PTRA, particularly if the angioplasty
to-control hypertension. Other clinical presentations of renal artery was inadequate. Most instances of restenosis can be successfully
FMD include flank pain and/or hematuria, both of which can result treated with repeat angioplasty. Use of intravascular ultrasound (IVUS)
from dissection or occlusion of the renal artery and aneurysmal in this setting can document resolution of a pressure gradient. Similar
changes involving the artery. Interestingly, unless dissection occurs, to patients on antihypertensive medications, patients are advised to
medial fibroplasia rarely results in renal dysfunction. Intimal and continue with routine surveillance for recurrence of hypertension
perimedial fibroplasia may be associated with renal dysfunction, and for Doppler ultrasound of renal length and cortical thickness.
arterial dissection, and progression to occlusion (Stokes et al., 1996). Routine use of aspirin (81 mg) is advised for all patients with renal
Diagnostic Evaluation. As with atherosclerotic renal artery stenosis, artery FMD (Olin, 2011).
angiography remains the gold standard to diagnose and evaluate
renal artery FMD. Catheter-based angiography provides excellent
resolution of the main and segmental renal arteries and can identify KEY POINTS: CHRONIC KIDNEY DIEASE
luminal changes such as aneurysm formation and dissection that • CKD is defined as kidney impairment sustained beyond 3
may affect the smaller vessels. More recently intravascular ultrasound months with a reduction in GFR, structural abnormalities
has been used as an adjunct to angiography and can depict pressure in the kidneys, and/or proteinuria.
• There continues to be a steady increase in the number of
patients with ESRD on RRTs throughout the world.
Outcome comparisons suggest that kidney transplantation
is still the best overall treatment for ESRD patients, despite
advances in dialysis care and modalities.
• Hemodynamically significant narrowing in the renal artery,
or renal artery stenosis, can result in secondary
hypertension and/or diminishing renal function.
• Atherosclerotic vascular disease accounts for 90% of causes
of renal artery stenosis, and fibromuscular dysplasias
account for the remaining 10%.
• Renal artery stenosis can lead to renal parenchymal loss
and diminished renal function, a condition known as
ischemic nephropathy.
• Although the limitations of these studies continue to be
debated, the conclusions reached by the CORAL study
reliably show the lack of benefit of renal artery stenting in
Fig. 87.5. “String of beads” appearance of the right renal artery typical of most patients with atherosclerotic renal artery stenosis.
fibromuscular dysplasias.

Downloaded for FK UMI Makassar (mahasiswafkumi09@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier on January 10, 2021.
For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Chapter 87 Renal Insufficiency and Ischemic Nephropathy 1935

SUGGESTED READINGS Goicoechea M, Rivera F, López-Gómez JM: Increased prevalence of acute


tubulointerstitial nephritis, Nephrol Dial Transplant 28(1):112–115, 2013.
Bax L, Woittiez AJ, Kouwenberg HJ, et al: Stent placement in patients with Khosroshahi A, Wallace ZS, Crowe JL, et al: International consensus guidance
atherosclerotic renal artery stenosis and impaired renal function: a random- statement on the management and treatment of IgG4-related disease,
ized trial, Ann Intern Med 150(12):840–848, W150–841, 2009. Arthritis Rheumatol 67(7):1688–1699, 2015.
Beck LH, Bonegio RG, Lambeau G, et al: M-type phospholipase A2 receptor Liaño F, Pascual J: Epidemiology of acute renal failure: a prospective, mul-
as target antigen in idiopathic membranous nephropathy, N Engl J Med ticenter, community-based study. Madrid Acute Renal Failure Study Group,
361(1):11–21, 2009. Kidney Int 50(3):811–818, 1996.
Cavanaugh C, Perazella MA: Urine sediment examination in the diagnosis May AG, De Weese JA, Rob CG: Hemodynamic effects of arterial stenosis,
and management of kidney disease: core curriculum 2019, Am J Kidney Surgery 53:513–524, 1963.
Dis 2018.
Chawla LS, Kimmel PL: Acute kidney injury and chronic kidney disease: an
integrated clinical syndrome, Kidney Int 82(5):516–524, 2012.
Cooper CJ, Murphy TP, Cutlip DE, et al: Stenting and medical therapy for
REFERENCES
atherosclerotic renal-artery stenosis, N Engl J Med 370(1):13–22, 2014. The complete reference list is available online at ExpertConsult.com.
Freedman BI, Limou S, Ma L, et al: APOL1-Associated Nephropathy: a key
contributor to racial disparities in CKD, Am J Kidney Dis 72(5S1):S8–S16,
2018.

Downloaded for FK UMI Makassar (mahasiswafkumi09@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier on January 10, 2021.
For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Chapter 87 Renal Insufficiency and Ischemic Nephropathy 1935.e1

Chade AR, Rodriguez-Porcel M, Grande JP, et al: Mechanisms of renal structural


REFERENCES alterations in combined hypercholesterolemia and renal artery stenosis,
AbuRahma AF, Srivastava M, Mousa AY, et al: Critical analysis of renal duplex Arterioscler Thromb Vasc Biol 23(7):1295–1301, 2003.
ultrasound parameters in detecting significant renal artery stenosis, J Vasc Chawla LS, Kimmel PL: Acute kidney injury and chronic kidney disease: an
Surg 56(4):1052–1059, 1060 e1051; discussion 1059–1060, 2012. integrated clinical syndrome, Kidney Int 82(5):516–524, 2012.
Anema JG, Morey AF, McAninch JW, et al: Complications related to the high Cherr GS, Hansen KJ, Craven TE, et al: Surgical management of atherosclerotic
lithotomy position during urethral reconstruction, J Urol 164(2):360–363, 2000. renovascular disease, J Vasc Surg 35(2):236–245, 2002.
Angeli P, Volpin R, Gerunda G, et al: Reversal of type 1 hepatorenal syndrome Cohen SD, Kopp JB, Kimmel PL: Kidney diseases associated with human
with the administration of midodrine and octreotide, Hepatology immunodeficiency virus infection, N Engl J Med 377(24):2363–2374, 2017.
29(6):1690–1697, 1999. Coiffier B, Altman A, Pui CH, et al: Guidelines for the management of pediatric
Baker RJ, Pusey CD: The changing profile of acute tubulointerstitial nephritis, and adult tumor lysis syndrome: an evidence-based review, J Clin Oncol
Nephrol Dial Transplant 19(1):8–11, 2004. 26(16):2767–2778, 2008.
Balasubramanian G, Al-Aly Z, Moiz A, et al: Early nephrologist involvement Conlon PJ, Little MA, Pieper K, et al: Severity of renal vascular disease predicts
in hospital-acquired acute kidney injury: a pilot study, Am J Kidney Dis mortality in patients undergoing coronary angiography, Kidney Int
57(2):228–234, 2011. 60(4):1490–1497, 2001.
Bankir L, Bichet DG, Morgenthaler NG: Vasopressin: physiology, assessment Cooper CJ, Murphy TP, Cutlip DE, et al: Stenting and medical therapy for
and osmosensation, J Intern Med 282(4):284–297, 2017. atherosclerotic renal-artery stenosis, N Engl J Med 370(1):13–22, 2014.
Barbar SD, Clere-Jehl R, Bourredjem A, et al: Timing of renal-replacement Cortazar FB, Marrone KA, Troxell ML, et al: Clinicopathological features of
therapy in patients with acute kidney injury and sepsis, N Engl J Med acute kidney injury associated with immune checkpoint inhibitors, Kidney
379(15):1431–1442, 2018. Int 90(3):638–647, 2016.
Batlle D, Arruda J: Hyperkalemic forms of renal tubular acidosis: clinical and Cosentino F: Drugs for the prevention and treatment of acute renal failure,
pathophysiological aspects, Adv Chronic Kidney Dis 25(4):321–333, 2018. Cleve Clin J Med 62(4):248–253, 1995.
Bax L, Woittiez AJ, Kouwenberg HJ, et al: Stent placement in patients with Cragg AH, Smith TP, Thompson BH, et al: Incidental fibromuscular dysplasia
atherosclerotic renal artery stenosis and impaired renal function: a random- in potential renal donors: long-term clinical follow-up, Radiology
ized trial, Ann Intern Med 150(12):840–848, W150–841, 2009. 172(1):145–147, 1989.
Beck LH, Bonegio RG, Lambeau G, et al: M-type phospholipase A2 receptor Darling RC 3rd, Kreienberg PB, Chang BB, et al: Outcome of renal artery
as target antigen in idiopathic membranous nephropathy, N Engl J Med reconstruction: analysis of 687 procedures, Ann Surg 230(4):524–530,
361(1):11–21, 2009. discussion 530–522, 1999.
Beck LH, Fervenza FC, Beck DM, et al: Rituximab-induced depletion of Davies MG, Saad WE, Peden EK, et al: The long-term outcomes of percutaneous
anti-PLA2R autoantibodies predicts response in membranous nephropathy, therapy for renal artery fibromuscular dysplasia, J Vasc Surg 48(4):865–871,
J Am Soc Nephrol 22(8):1543–1550, 2011. 2008.
Bellomo R, Chapman M, Finfer S, et al: Low-dose dopamine in patients with De Bruyne B, Manoharan G, Pijls NH, et al: Assessment of renal artery stenosis
early renal dysfunction: a placebo-controlled randomised trial. Australian severity by pressure gradient measurements, J Am Coll Cardiol 48(9):1851–
and New Zealand Intensive Care Society (ANZICS) Clinical Trials Group, 1855, 2006.
Lancet 356(9248):2139–2143, 2000. de Mast Q, Beutler JJ: The prevalence of atherosclerotic renal artery stenosis in
Bellomo R, Ronco C, Kellum JA, et al., workgroup ADQI. Acute renal failure risk groups: a systematic literature review, J Hypertens 27(7):1333–1340, 2009.
- definition, outcome measures, animal models, fluid therapy and informa- De Vriese AS, Sethi S, Nath KA, et al: Differentiating primary, genetic, and
tion technology needs: the Second International Consensus Conference secondary FSGS in adults: a clinicopathologic approach, J Am Soc Nephrol
of the Acute Dialysis Quality Initiative (ADQI) Group, Crit Care 29(3):759–774, 2018.
8(4):R204–R212, 2004. Deane LA, Lee HJ, Box GN, et al: Third place: flank position is associated
Better OS, Arieff AI, Massry SG, et al: Studies on renal function after relief with higher skin-to-surface interface pressures in men versus women:
of complete unilateral ureteral obstruction of three months’ duration in implications for laparoscopic renal surgery and the risk of rhabdomyolysis,
man, Am J Med 54(2):234–240, 1973. J Endourol 22(6):1147–1151, 2008.
Bhathena DB, Julian BA, McMorrow RG, et al: Focal sclerosis of hypertrophied Doolan PD, Alpen EL, Theil GB: A clinical appraisal of the plasma concentra-
glomeruli in solitary functioning kidneys of humans, Am J Kidney Dis tion and endogenous clearance of creatinine, Am J Med 32:65–79, 1962.
5(5):226–232, 1985. Edwards MS, Corriere MA: Contemporary management of atherosclerotic
Bock JS, Gottlieb SS: Cardiorenal syndrome: new perspectives, Circulation renovascular disease, J Vasc Surg 50(5):1197–1210, 2009.
121(23):2592–2600, 2010. Edwards MS, Craven TE, Burke GL, et al: Renovascular disease and the risk
Bollée G, Patey N, Cazajous G, et al: Thrombotic microangiopathy secondary of adverse coronary events in the elderly: a prospective, population-based
to VEGF pathway inhibition by sunitinib, Nephrol Dial Transplant study, Arch Intern Med 165(2):207–213, 2005.
24(2):682–685, 2009. Eirin A, Gloviczki ML, Tang H, et al: Inflammatory and injury signals released
Bouquegneau A, Vidal-Petiot E, Moranne O, et al: Creatinine-based equations from the post-stenotic human kidney, Eur Heart J 34(7):540–548a, 2013.
for the adjustment of drug dosage in an obese population, Br J Clin Eklof H, Ahlstrom H, Magnusson A, et al: A prospective comparison of duplex
Pharmacol 81(2):349–361, 2016. ultrasonography, captopril renography, MRA, and CTA in assessing renal
Bove T, Zangrillo A, Guarracino F, et al: Effect of fenoldopam on use of renal artery stenosis, Acta Radiol 47(8):764–774, 2006.
replacement therapy among patients with acute kidney injury after cardiac Eng J, Wilson RF, Subramaniam RM, et al: Comparative effect of contrast
surgery: a randomized clinical trial, J Am Med Assoc 312(21):2244–2253, 2014. media type on the incidence of contrast-induced nephropathy: a systematic
Brenner BM, Mackenzie HS: Nephron mass as a risk factor for progression review and meta-analysis, Ann Intern Med 164(6):417–424, 2016.
of renal disease, Kidney Int Suppl 63:S124–S127, 1997. Eremina V, Jefferson JA, Kowalewska J, et al: VEGF inhibition and renal
Bushinsky DA, Williams GH, Pitt B, et al: Patiromer induces rapid and sustained thrombotic microangiopathy, N Engl J Med 358(11):1129–1136, 2008.
potassium lowering in patients with chronic kidney disease and hyperka- Fehrman-Ekholm I, Kvarnström N, Söfteland JM, et al: Post-nephrectomy
lemia, Kidney Int 88(6):1427–1433, 2015. development of renal function in living kidney donors: a cross-sectional
Cambria RP, Brewster DC, L’Italien GJ, et al: Renal artery reconstruction for retrospective study, Nephrol Dial Transplant 26(7):2377–2381, 2011.
the preservation of renal function, J Vasc Surg 24(3):371–380, discussion Felker GM, Lee KL, Bull DA, et al: Diuretic strategies in patients with acute
380–372, 1996. decompensated heart failure, N Engl J Med 364(9):797–805, 2011.
Caps MT, Zierler RE, Polissar NL, et al: Risk of atrophy in kidneys with Fergany A, Kolettis P, Novick AC: The contemporary role of extra-anatomical
atherosclerotic renal artery stenosis, Kidney Int 53(3):735–742, 1998. surgical renal revascularization in patients with atherosclerotic renal artery
Carvounis CP, Nisar S, Guro-Razuman S: Significance of the fractional excretion disease, J Urol 153(6):1798–1801, discussion 1801–1792, 1995.
of urea in the differential diagnosis of acute renal failure, Kidney Int Fordjour KN, Walton T, Doran JJ: Management of hyperkalemia in hospitalized
62(6):2223–2229, 2002. patients, Am J Med Sci 347(2):93–100, 2014.
Cavallin M, Kamath PS, Merli M, et al: Terlipressin plus albumin versus Formica RN, Aeder M, Boyle G, et al: Simultaneous liver-kidney allocation
midodrine and octreotide plus albumin in the treatment of hepatorenal policy: a proposal to optimize appropriate utilization of scarce resources,
syndrome: a randomized trial, Hepatology 62(2):567–574, 2015. Am J Transplant 16(3):758–766, 2016.
Cavanaugh C, Perazella MA: Urine sediment examination in the diagnosis Freedman BI, Limou S, Ma L, et al: APOL1-associated nephropathy: a key
and management of kidney disease: core curriculum 2019, Am J Kidney contributor to racial disparities in CKD, Am J Kidney Dis 72(5S1):S8–S16,
Dis 2018. 2018.
Chabova V, Schirger A, Stanson AW, et al: Outcomes of atherosclerotic Freedman BI, Pastan SO, Israni AK, et al: APOL1 genotype and kidney
renal artery stenosis managed without revascularization, Mayo Clin Proc transplantation outcomes from deceased African American donors,
75(5):437–444, 2000. Transplantation 100(1):194–202, 2016.

Downloaded for FK UMI Makassar (mahasiswafkumi09@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier on January 10, 2021.
For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
1935.e2 PART VIII Renal Physiology and Pathophysiology

Friedman DJ, Kozlitina J, Genovese G, et al: Population-based risk assessment Keene R, Froelich JM, Milbrandt JC, et al: Bilateral gluteal compartment
of APOL1 on renal disease, J Am Soc Nephrol 22(11):2098–2105, 2011. syndrome following robotic-assisted prostatectomy, Orthopedics 33(11):852,
Fulton C, McGregor T, Forbes TL, et al: Catheter-directed thrombolysis with 2010.
tPA to restore renal function after iliac venous thrombosis post-renal Khosroshahi A, Wallace ZS, Crowe JL, et al: International consensus guidance
transplantation, Ann Transplant 16(1):61–65, 2011. statement on the management and treatment of IgG4-related disease,
Geevasinga N, Coleman PL, Webster AC, et al: Proton pump inhibitors and Arthritis Rheumatol 67(7):1688–1699, 2015.
acute interstitial nephritis, Clin Gastroenterol Hepatol 4(5):597–604, 2006. Kidney Disease: Improving Global outcomes section 2: AKI definition, Kidney
Giacoppo D, Madhavan MV, Baber U, et al: Impact of contrast-induced acute Int Suppl (2011) 2(1):19–36, 2012.
kidney injury after percutaneous coronary intervention on short- and Kim HS, Fine DM, Atta MG: Catheter-directed thrombectomy and thrombolysis
long-term outcomes: pooled analysis from the HORIZONS-AMI and ACUITY for acute renal vein thrombosis, J Vasc Interv Radiol 17(5):815–822, 2006.
Trials, Circ Cardiovasc Interv 8(8):e002475, 2015. Klahr S: New insights into the consequences and mechanisms of renal
Glassman DT, Merriam WG, Trabulsi EJ, et al: Rhabdomyolysis after laparo- impairment in obstructive nephropathy, Am J Kidney Dis 18(6):689–699,
scopic nephrectomy, JSLS 11(4):432–437, 2007. 1991.
Gloviczki ML, Saad A, Textor SC: Blood oxygen level-dependent (BOLD) Knight EL, Verhave JC, Spiegelman D, et al: Factors influencing serum cystatin
MRI analysis in atherosclerotic renal artery stenosis, Curr Opin Nephrol C levels other than renal function and the impact on renal function
Hypertens 22(5):519–524, 2013. measurement, Kidney Int 65(4):1416–1421, 2004.
Goicoechea M, Rivera F, López-Gómez JM: Increased prevalence of acute Konstam MA, Gheorghiade M, Burnett JC, et al: Effects of oral tolvaptan in
tubulointerstitial nephritis, Nephrol Dial Transplant 28(1):112–115, 2013. patients hospitalized for worsening heart failure: the EVEREST Outcome
Goldblatt H, Weinstein H, Kahn JR: Studies on experimental hypertension Trial, J Am Med Assoc 297(12):1319–1331, 2007.
: xiv. The effect of intermittent renal arterial occlusion on the blood pressure Kuang W, Ng CS, Matin S, et al: Rhabdomyolysis after laparoscopic donor
of the dog, J Exper Med 73(3):439–451, 1941. nephrectomy, Urology 60(5):911, 2002.
Gonwa TA, Morris CA, Goldstein RM, et al: Long-term survival and renal func- Kumbhani DJ, Bavry AA, Harvey JE, et al: Clinical outcomes after percutaneous
tion following liver transplantation in patients with and without hepatorenal revascularization versus medical management in patients with significant
syndrome–experience in 300 patients, Transplantation 51(2):428–430, 1991. renal artery stenosis: a meta-analysis of randomized controlled trials, Am
Gottlieb RH, Weinberg EP, Rubens DJ, et al: Renal sonography: can it be Heart J 161(3):622–630, e621, 2011.
used more selectively in the setting of an elevated serum creatinine level?, Larsen CP, Beggs ML, Saeed M, et al: Apolipoprotein L1 risk variants associate
Am J Kidney Dis 29(3):362–367, 1997. with systemic lupus erythematosus-associated collapsing glomerulopathy,
Goumenos DS, Kawar B, El Nahas M, et al: Early histological changes in the J Am Soc Nephrol 24(5):722–725, 2013.
kidney of people with morbid obesity, Nephrol Dial Transplant 24(12):3732– Lautin EM, Freeman NJ, Schoenfeld AH, et al: Radiocontrast-associated renal
3738, 2009. dysfunction: a comparison of lower-osmolality and conventional high-
Goutorbe P, Montcriol A, Lacroix G, et al: Intestinal necrosis associated with osmolality contrast media, Am J Roentgenol 157(1):59–65, 1991.
orally administered calcium polystyrene sulfonate without sorbitol, Ann Lazarus B, Chen Y, Wilson FP, et al: Proton pump inhibitor use and the risk
Pharmacother 45(2):e13, 2011. of chronic kidney disease, J Am Med Assoc Intern Med 176(2):238–246,
Hagan AE, Farrington CA, Wall GC, et al: Sodium polystyrene sulfonate for 2016.
the treatment of acute hyperkalemia: a retrospective study, Clin Nephrol Lerman LO, Nath KA, Rodriguez-Porcel M, et al: Increased oxidative stress
85(1):38–43, 2016. in experimental renovascular hypertension, Hypertension 37(2 Pt 2):541–546,
Hansen KJ, Cherr GS, Craven TE, et al: Management of ischemic nephropathy: 2001.
dialysis-free survival after surgical repair, J Vasc Surg 32(3):472–481, discus- Lerman LO, Textor SC, Grande JP: Mechanisms of tissue injury in renal artery
sion 481–472, 2000. stenosis: ischemia and beyond, Prog Cardiovasc Dis 52(3):196–203, 2009.
Hansen KJ, Edwards MS, Craven TE, et al: Prevalence of renovascular disease Leung N, Bridoux F, Hutchison CA, et al: Monoclonal gammopathy of renal
in the elderly: a population-based study, J Vasc Surg 36(3):443–451, 2002. significance: when MGUS is no longer undetermined or insignificant,
Heart Outcomes Prevention Evaluation Study Investigators: Effects of Ramipril Blood 120(22):4292–4295, 2012.
on cardiovascular and microvascular outcomes in people with diabetes Levey AS, Bosch JP, Lewis JB, et al: A more accurate method to estimate
mellitus: results of the HOPE study and MICRO-HOPE substudy, Lancet glomerular filtration rate from serum creatinine: a new prediction equation.
355(9200):253–259, 2000. Modification of Diet in Renal Disease Study Group, Ann Intern Med
Holley KE, Hunt JC, Brown AL Jr, et al: Renal artery stenosis. A clinical- 130(6):461–470, 1999.
pathologic study in normotensive and hypertensive patients, Am J Med Levey AS, Stevens LA, Schmid CH, et al: A new equation to estimate glomerular
37:14–22, 1964. filtration rate, Ann Intern Med 150(9):604–612, 2009.
Hoorn EJ, Tuut MK, Hoorntje SJ, et al: Dutch guideline for the management Lewington AJ, Cerdá J, Mehta RL: Raising awareness of acute kidney injury:
of electrolyte disorders–2012 revision, Neth J Med 71(3):153–165, 2013. a global perspective of a silent killer, Kidney Int 84(3):457–467, 2013.
Howard SC, Jones DP, Pui CH: The tumor lysis syndrome, N Engl J Med Lewis SJ, Mueller BA: Antibiotic dosing in patients with acute kidney injury:
364(19):1844–1854, 2011. “enough but not too much”, J Intens Care Med 31(3):164–176, 2016.
Hoxha E, Wiech T, Stahl PR, et al: A mechanism for cancer-associated Liang P, Hurks R, Bensley RP, et al: The rise and fall of renal artery angioplasty
membranous nephropathy, N Engl J Med 374(20):1995–1996, 2016. and stenting in the United States, 1988-2009, J Vasc Surg 58(5):1331–1338,
Hoy SM: Sodium zirconium cyclosilicate: a review in hyperkalaemia, Drugs e1331, 2013.
78(15):1605–1613, 2018. Liaño F, Pascual J: Epidemiology of acute renal failure: a prospective, mul-
Hughson M, Farris AB, Douglas-Denton R, et al: Glomerular number and ticenter, community-based study. Madrid Acute Renal Failure Study Group,
size in autopsy kidneys: the relationship to birth weight, Kidney Int Kidney Int 50(3):811–818, 1996.
63(6):2113–2122, 2003. Lin CJ, Lim KH, Cheng YC, et al: Tumor lysis syndrome after treatment with
Inker LA, Schmid CH, Tighiouart H, et al: Estimating glomerular filtration rate gemcitabine for metastatic transitional cell carcinoma, Med Oncol
from serum creatinine and cystatin C, N Engl J Med 367(1):20–29, 2012. 24(4):455–457, 2007.
Iwakiri Y: The molecules: mechanisms of arterial vasodilatation observed in Lopes AA, Port FK: The low birth weight hypothesis as a plausible explanation
the splanchnic and systemic circulation in portal hypertension, J Clin for the black/white differences in hypertension, non-insulin-dependent
Gastroenterol 41(Suppl 3):S288–S294, 2007. diabetes, and end-stage renal disease, Am J Kidney Dis 25(2):350–356,
Izzedine H, Mateus C, Boutros C, et al: Renal effects of immune checkpoint 1995.
inhibitors, Nephrol Dial Transplant 32(6):936–942, 2017. Loscalzo J: Venous thrombosis in the nephrotic syndrome, N Engl J Med
Johansson M, Herlitz H, Jensen G, et al: Increased cardiovascular mortality 368(10):956–958, 2013.
in hypertensive patients with renal artery stenosis. Relation to sympathetic Luo Y, Wang X, Ye Z, et al: Remedial hydration reduces the incidence of
activation, renal function and treatment regimens, J Hypertens 17(12 Pt contrast-induced nephropathy and short-term adverse events in patients
1):1743–1750, 1999. with ST-segment elevation myocardial infarction: a single-center, randomized
Julian BA, Gaston RS, Brown WM, et al: Effect of replacing race with apoli- trial, Intern Med 53(20):2265–2272, 2014.
poprotein L1 genotype in calculation of kidney donor risk index, Am J Lye WC, Cheah JS, Sinniah R: Renal cholesterol embolic disease. Case report
Transplant 17(6):1540–1548, 2017. and review of the literature, Am J Nephrol 13(6):489–493, 1993.
Jurado-Román A, Hernández-Hernández F, García-Tejada J, et al: Role of Madhavan SM, O’Toole JF, Konieczkowski M, et al: APOL1 localization in
hydration in contrast-induced nephropathy in patients who underwent normal kidney and nondiabetic kidney disease, J Am Soc Nephrol
primary percutaneous coronary intervention, Am J Cardiol 115(9):1174–1178, 22(11):2119–2128, 2011.
2015. Mailloux LU, Kapikian N, Napolitano B, et al: Home hemodialysis: patient
Kamholtz RG, Cronan JJ, Dorfman GS: Obstruction and the minimally dilated outcomes during a 24-year period of time from 1970 through 1993, Adv
renal collecting system: US evaluation, Radiology 170(1 Pt 1):51–53, 1989. Ren Replace Ther 3(2):112–119, 1996.

Downloaded for FK UMI Makassar (mahasiswafkumi09@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier on January 10, 2021.
For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
Chapter 87 Renal Insufficiency and Ischemic Nephropathy 1935.e3

Manetti L, Pardini E, Genovesi M, et al: Thyroid function differently affects Ren S, Wu C, Zhang Y, et al: An update on clinical significance of use of
serum cystatin C and creatinine concentrations, J Endocrinol Invest THSD7A in diagnosing idiopathic membranous nephropathy: a systematic
28(4):346–349, 2005. review and meta-analysis of THSD7A in IMN, Ren Fail 40(1):306–313,
Marone LK, Clouse WD, Dorer DJ, et al: Preservation of renal function with 2018.
surgical revascularization in patients with atherosclerotic renovascular Rich MW, Crecelius CA: Incidence, risk factors, and clinical course of acute
disease, J Vasc Surg 39(2):322–329, 2004. renal insufficiency after cardiac catheterization in patients 70 years of age
Matsushita K, Mahmoodi BK, Woodward M, et al: Comparison of risk prediction or older. A prospective study, Arch Intern Med 150(6):1237–1242, 1990.
using the CKD-EPI equation and the MDRD study equation for estimated Ritchie J, Green D, Chrysochou C, et al: High-risk clinical presentations in
glomerular filtration rate, J Am Med Assoc 307(18):1941–1951, 2012. atherosclerotic renovascular disease: prognosis and response to renal artery
May AG, De Weese JA, Rob CG: Hemodynamic effects of arterial stenosis, revascularization, Am J Kidney Dis 63(2):186–197, 2014.
Surgery 53:513–524, 1963. Rocha-Singh KJ, Eisenhauer AC, Textor SC, et al: Atherosclerotic Peripheral
McGregor DO, Buttimore AL, Lynn KL: Home hemodialysis: excellent survival Vascular Disease Symposium II: intervention for renal artery disease, Circula-
at less cost, but still underutilized, Kidney Int 57(6):2654–2655, 2000. tion 118(25):2873–2878, 2008.
McKenzie GA, Oderich GS, Kawashima A, et al: Renal artery fibromuscular Ruggenenti P, Debiec H, Ruggiero B, et al: Anti-phospholipase a2 receptor
dysplasia in 2,640 renal donor subjects: a CT angiography analysis, J Vasc antibody titer predicts post-rituximab outcome of membranous nephropathy,
Interv Radiol 24(10):1477–1480, 2013. J Am Soc Nephrol 26(10):2545–2558, 2015.
Mehta RL, McDonald B, Gabbai F, et al: Nephrology consultation in acute Sadowski CE, Lovric S, Ashraf S, et al: A single-gene cause in 29.5% of cases
renal failure: does timing matter?, Am J Med 113(6):456–461, 2002. of steroid-resistant nephrotic syndrome, J Am Soc Nephrol 26(6):1279–1289,
Meier-Kriesche HU, Port FK, Ojo AO, et al: Effect of waiting time on renal 2015.
transplant outcome, Kidney Int 58(3):1311–1317, 2000. Saeki T, Nishi S, Imai N, et al: Clinicopathological characteristics of patients
Miralles M, Corominas A, Cotillas J, et al: Screening for carotid and renal with IgG4-related tubulointerstitial nephritis, Kidney Int 78(10):1016–1023,
artery stenoses in patients with aortoiliac disease, Ann Vasc Surg 12(1):17–22, 2010.
1998. Schetz M, Gunst J, Van den Berghe G: The impact of using estimated GFR
Muriithi AK, Nasr SH, Leung N: Utility of urine eosinophils in the diagnosis versus creatinine clearance on the evaluation of recovery from acute kidney
of acute interstitial nephritis, Clin J Am Soc Nephrol 8(11):1857–1862, injury in the ICU, Intensive Care Med 40(11):1709–1717, 2014.
2013. Schlossberg SM, Vaughan ED: The mechanism of unilateral post-obstructive
Myers BD, Moran SM: Hemodynamically mediated acute renal failure, N diuresis, J Urol 131(3):534–536, 1984.
Engl J Med 314(2):97–105, 1986. Schnermann J, Homer W: Smith Award lecture. The juxtaglomerular apparatus:
Newman DJ, Thakkar H, Edwards RG, et al: Serum cystatin C measured by from anatomical peculiarity to physiological relevance, J Am Soc Nephrol
automated immunoassay: a more sensitive marker of changes in GFR than 14(6):1681–1694, 2003.
serum creatinine, Kidney Int 47(1):312–318, 1995. Schold JD, Sehgal AR, Srinivas TR, et al: Marked variation of the association
Neymark E, LaBerge JM, Hirose R, et al: Arteriographic detection of renovascular of ESRD duration before and after wait listing on kidney transplant
disease in potential renal donors: incidence and effect on donor surgery, outcomes, Am J Transplant 10(9):2008–2016, 2010.
Radiology 214(3):755–760, 2000. Schwab SJ, Hlatky MA, Pieper KS, et al: Contrast nephrotoxicity: a randomized
Norberg SM, Oros M, Birkenbach M, et al: Spontaneous tumor lysis syndrome controlled trial of a nonionic and an ionic radiographic contrast agent,
in renal cell carcinoma: a case report, Clin Genitourin Cancer 12(5):e225– N Engl J Med 320(3):149–153, 1989.
e227, 2014. Schwartz CJ, White TA: Stenosis of renal artery: an unselected necropsy study,
Novick AC, Gephardt G, Guz B, et al: Long-term follow-up after partial removal Br Med J 2(5422):1415–1421, 1964.
of a solitary kidney, N Engl J Med 325(15):1058–1062, 1991. Scolari F, Ravani P, Gaggi R, et al: The challenge of diagnosing atheroembolic
Novick AC, Schreiber MJ: Effect of angiotensin-converting enzyme inhibition renal disease: clinical features and prognostic factors, Circulation
on nephropathy in patients with a remnant kidney, Urology 46(6):785–789, 116(3):298–304, 2007.
1995. Sethi S, Rajkumar SV: Monoclonal gammopathy-associated proliferative
Olin JW, Sealove BA: Diagnosis, management, and future developments of glomerulonephritis, Mayo Clin Proc 88(11):1284–1293, 2013.
fibromuscular dysplasia, J Vasc Surg 53(3):826–836, e821, 2011. Skluzacek PA, Szewc RG, Nolan CR 3rd, et al: Prediction of GFR in liver
Pannu N: Bidirectional relationships between acute kidney injury and chronic transplant candidates, Am J Kidney Dis 42(6):1169–1176, 2003.
kidney disease, Curr Opin Nephrol Hypertens 22(3):351–356, 2013. Somerville CA, Chmiel R, Niall JF, et al: Non-dilated urinary tract obstruction,
Parsa A, Kao WH, Xie D, et al: APOL1 risk variants, race, and progression of Med J Aust 156(10):721–723, 1992.
chronic kidney disease, N Engl J Med 369(23):2183–2196, 2013. Stevens LA, Levey AS: Measured GFR as a confirmatory test for estimated
Paty PS, Darling RC 3rd, Lee D, et al: Is prosthetic renal artery reconstruction GFR, J Am Soc Nephrol 20(11):2305–2313, 2009.
a durable procedure? An analysis of 489 bypass grafts, J Vasc Surg Stokes JB, Bonsib SM, McBride JW: Diffuse intimal fibromuscular dysplasia
34(1):127–132, 2001. with multiorgan failure, Arch Intern Med 156(22):2611–2614, 1996.
Pépin MN, Bouchard J, Legault L, et al: Diagnostic performance of fractional Testani JM, Khera AV, St John Sutton MG, et al: Effect of right ventricular
excretion of urea and fractional excretion of sodium in the evaluations function and venous congestion on cardiorenal interactions during the
of patients with acute kidney injury with or without diuretic treatment, treatment of decompensated heart failure, Am J Cardiol 105(4):511–516,
Am J Kidney Dis 50(4):566–573, 2007. 2010.
Perazella MA, Parikh CR: How can urine microscopy influence the differential Trinquart L, Mounier-Vehier C, Sapoval M, et al: Efficacy of revascularization
diagnosis of AKI?, Clin J Am Soc Nephrol 4(4):691–693, 2009. for renal artery stenosis caused by fibromuscular dysplasia: a systematic
Persson PB, Hansell P, Liss P: Pathophysiology of contrast medium-induced review and meta-analysis, Hypertension 56(3):525–532, 2010.
nephropathy, Kidney Int 68(1):14–22, 2005. U.S. Renal Data System. USRDS 2013 Annual Data Report: Atlas of Chronic
Pfister SA, Deckart A, Laschke S, et al: Unenhanced helical computed Kidney Disease and End Stage Renal Disease in the United States. Bethesda,
tomography vs intravenous urography in patients with acute flank pain: MD: National Institutes of Health, National Institute of Diabetes and
accuracy and economic impact in a randomized prospective trial, Eur Digestive and Kidney Diseases, 2013.
Radiol 13(11):2513–2520, 2003. US Renal Data System. USRDS 2013 Annual Data Report: Epidemiology of
Plouin PF, Chatellier G, Darne B, et al: Blood pressure outcome of angioplasty Kidney Disease in the United States. Bethesda, MD: National Institutes of
in atherosclerotic renal artery stenosis: a randomized trial. Essai Multi- Health, National Institute of Diabetes and Digestive and Kidney Diseases,
centrique Medicaments vs Angioplastie (EMMA) Study Group, Hypertension 2013.
31(3):823–829, 1998. Uchino S, Kellum JA, Bellomo R, et al: Acute renal failure in critically ill
Pöge U, Gerhardt T, Stoffel-Wagner B, et al: Calculation of glomerular filtration patients: a multinational, multicenter study, J Am Med Assoc 294(7):813–818,
rate based on cystatin C in cirrhotic patients, Nephrol Dial Transplant 2005.
21(3):660–664, 2006. van de Ven PJ, Kaatee R, Beutler JJ, et al: Arterial stenting and balloon
Ponce D, Zorzenon Cde P, dos Santos NY, et al: Early nephrology consultation angioplasty in ostial atherosclerotic renovascular disease: a randomised
can have an impact on outcome of acute kidney injury patients, Nephrol trial, Lancet 353(9149):282–286, 1999.
Dial Transplant 26(10):3202–3206, 2011. van Jaarsveld BC, Krijnen P, Pieterman H, et al: The effect of balloon angioplasty
Prasad A, Zafar N, Mahmud E: Assessment of renal artery fibromuscular on hypertension in atherosclerotic renal-artery stenosis. Dutch Renal Artery
dysplasia: angiography, intravascular ultrasound (with virtual histology), Stenosis Intervention Cooperative Study Group, N Engl J Med 342(14):1007–
and pressure wire measurements, Catheter Cardiovasc Interv 74(2):260–264, 1014, 2000.
2009. Vanhamme L, Paturiaux-Hanocq F, Poelvoorde P, et al: Apolipoprotein L-I
Prowle JR, Kirwan CJ, Bellomo R: Fluid management for the prevention and is the trypanosome lytic factor of human serum, Nature 422(6927):83–87,
attenuation of acute kidney injury, Nat Rev Nephrol 10(1):37–47, 2014. 2003.

Downloaded for FK UMI Makassar (mahasiswafkumi09@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier on January 10, 2021.
For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.
1935.e4 PART VIII Renal Physiology and Pathophysiology

Vasbinder GB, Nelemans PJ, Kessels AG, et al: Diagnostic tests for renal artery Weir MR, Bakris GL, Bushinsky DA, et al: Patiromer in patients with kidney
stenosis in patients suspected of having renovascular hypertension: a disease and hyperkalemia receiving RAAS inhibitors, N Engl J Med
meta-analysis, Ann Intern Med 135(6):401–411, 2001. 372(3):211–221, 2015.
Vijay MK, Vijay P, Kundu AK: Rhabdomyolysis and myogloginuric acute renal Weisbord SD, Gallagher M, Jneid H, et al: Outcomes after angiography with
failure in the lithotomy/exaggerated lithotomy position of urogenital sodium bicarbonate and acetylcysteine, N Engl J Med 378(7):603–614,
surgeries, Urol Ann 3(3):147–150, 2011. 2018.
Walsh M, Devereaux PJ, Garg AX, et al: Relationship between intraoperative Wheatley K, Ives N, et al: Revascularization versus medical therapy for renal-
mean arterial pressure and clinical outcomes after noncardiac surgery: artery stenosis, N Engl J Med 361(20):1953–1962, 2009.
toward an empirical definition of hypotension, Anesthesiology 119(3):507–515, Whelton A: Nephrotoxicity of nonsteroidal anti-inflammatory drugs: physiologic
2013. foundations and clinical implications, Am J Med 106(5B):13S–24S, 1999.
Webster J, Marshall F, Abdalla M, et al: Randomised comparison of percutane- Wu CC, Yeung LK, Tsai WS, et al: Incidence and factors predictive of acute
ous angioplasty vs continued medical therapy for hypertensive patients renal failure in patients with advanced liver cirrhosis, Clin Nephrol
with atheromatous renal artery stenosis. Scottish and Newcastle Renal 65(1):28–33, 2006.
Artery Stenosis Collaborative Group, J Hum Hypertens 12(5):329–335, Xie Y, Bowe B, Li T, et al: Proton Pump Inhibitors and Risk of Incident CKD
1998. and Progression to ESRD, J Am Soc Nephrol 27(10):3153–3163, 2016.

Downloaded for FK UMI Makassar (mahasiswafkumi09@gmail.com) at University of Muslim Indonesia from ClinicalKey.com by Elsevier on January 10, 2021.
For personal use only. No other uses without permission. Copyright ©2021. Elsevier Inc. All rights reserved.

You might also like