Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

AKDH

Etiology and Management of Edema: A Review


Abbal Koirala, Negiin Pourafshar, Arvin Daneshmand, Christopher S. Wilcox,
Sai Sudha Mannemuddhu, and Nayan Arora

The development of peripheral edema can often pose a significant diagnostic and therapeutic challenge for practitioners due to
its association with a wide variety of underlying disorders ranging in severity. Updates to the original Starling’s principle have
provided new mechanistic insights into edema formation. Additionally, contemporary data highlighting the role of hypochlor-
emia in the development of diuretic resistance provide a possible new therapeutic target. This article reviews the pathophys-
iology of edema formation and discusses implications for treatment.
Q 2022 by the National Kidney Foundation, Inc. All rights reserved.
Key Words: Edema, Heart failure, Diuretics, Oncotic pressures, Albumin

T otal body water is calculated as the percentage of the


lean body weight in adults (50% in women and 60%
in men).1 For a 70-kg adult, this would be 42 L. Two-
stream to the interstitium, a process commonly referred to
as capillary filtration. The interstitial fluid drains into the
lymphatics, ultimately returning fluid to the systemic circu-
third of the total body water is intracellular (28 L in a 70- lation. The flow into the lymphatics is the only route through
kg adult), and one-third is extracellular (14 L in a 70-kg which plasma proteins, generally filtered through the capil-
adult).2 Three-fourth of extracellular fluid is in the intersti- laries, are returned to the blood circulation.
tium (10.5 L in a 70-kg adult), and the rest is in plasma Alterations in the forces across the vascular endothelial
(3.5 L in a 70-kg adult)2 (Table 1). cells, alteration of the capillary surface area, and perme-
ability characteristics of the capillary wall regulate transca-
ETIOLOGY AND MANAGEMENT OF EDEMA pillary fluid flow, favoring fluid movement from the
Classically, edema is defined as the expansion of interstitial vascular space into the interstitium, resulting in edema.
volume resulting in palpable swelling. However, this defi- Edema does not become clinically apparent until the inter-
nition can be broadened to include fluid accumulation stitial volume increases by at least 2.5 to 3 L. Therefore, the
within cells (cellular edema) or the interstitial space (inter- initial movement of fluid from the vascular space into the
stitial edema). Several clinical conditions are associated interstitium cannot sustain edema, as the reduction in
with edema development, including heart failure, plasma volume would reduce tissue perfusion, resulting
cirrhosis, and nephrotic syndrome. in shock. Activation of the renin-angiotensin-aldosterone
Causes of Edema3 system (RAAS) results in the retention of sodium and wa-
ter by the kidneys to restore tissue perfusion. While some
PATHOPHYSIOLOGY OF EDEMA FORMATION of this fluid remains in the vascular space, much of it
Starling’s forces (hydrostatic and oncotic pressures) operate passes into the interstitial space, resulting in the mainte-
across the microvascular walls between the vascular and nance of edema.
interstitial compartments and are responsible for fluid and
solute exchange between the 2 compartments.4,5 Usually, Capillary Hemodynamics
the balance of forces acting across the microvascular walls The exchange of fluid between plasma and interstitium
of the capillaries favors the net flux of fluid from the blood- has been traditionally described by Starling’s law (Fig 1).

Net filtration ¼ Kf 3 ðDelta hydrostatic pressure 2 Delta oncotic pressureÞ


¼ Kf 3 ½ðPc 2 PisÞ 2 sðpp 2 pisÞ

From the Division of Nephrology, University of Washington, Seattle, WA


(A.K., N.A.); Division of Nephrology, MedStar Georgetown University Hospi-
tal, Washington DC (N.P., C.W., A.D.); and Divison of Nephrology, University where Kf is the capillary filtration coefficient (surface area available
of Tennessee, Knoxville, TN (S.M.) for fluid movement 3 hydraulic permeability); Pc and Pis are the
Financial Disclosures: The authors declare that they have no relevant finan- capillary and interstitial fluid hydrostatic pressures, respectively;
cial interests. pp and pis are the capillary and interstitial fluid oncotic pressures,
Address correspondence to Nayan Arora, MD, Division of Nephrology, Uni-
versity of Washington, 1959 NE Pacific Street Box 356521, Seattle, WA 98195.
respectively; and s represents the reflection coefficient of proteins
E-mail: narora@uw.edu across the capillary wall (with values ranging from 0 if completely
Ó 2022 by the National Kidney Foundation, Inc. All rights reserved. permeable to 1 if completely impermeable).
2949-8139/$36.00 Starling’s forces originally postulated fluid filtration at
https://doi.org/10.1053/j.akdh.2022.12.002 the arterial end and fluid resorption at the venular end

110 Adv in Kidney Disease and Health 2023;30(2):110-123


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en marzo 06, 2023. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
Etiology and Management of Edema: A Review 111

of the capillary bed. While this hypothesis is valid in the tion of the sympathetic and renin-angiotensin systems.
kidney cortex and medulla, net filtration continues These neurohumoral activations promote sodium and
throughout the capillary length elsewhere, invalidating water retention, leading to plasma volume expansion.
several assumptions of the Starling relationship.6,7 In a chronic compensated state of heart failure, the resul-
Albumin and sodium ions bound to glycosaminoglycans tant plasma volume expansion leads to the restoration of
in the interstitium contribute to pis and fluid filtration. cardiac output at the expense of an increase in LVEDP. A
Interstitial fluid albumin is recycled back to the systemic new steady state is established in which the stroke vol-
circulation through the lymphatics. The filtration barrier ume and cardiac output are normal, sodium excretion
is formed by a glycocalyx barrier that lines the capillary lu- matches sodium intake, and the activity of the RAAS
mens. The filtration barrier is interrupted by clefts through has returned to normal.9 Decompensation of this new
which filtration occurs. The oncotic pressure difference
7
steady state can occur from increased dietary sodium
across plasma-sub glycocalyx opposes filtration but does load,10 resulting in edema. Additionally, cardiac output
not reverse the filtration rate. Fluid filtration occurs may not be augmented further to support exercise in a
throughout the capillary wall from the arteriolar to the well-compensated state, resulting in decreased tissue
venular end. With the subacute reduction in capillary pres- perfusion and further neurohumoral activation, renal
sure, the glycocalyx model preserves filtration at a very vasoconstriction, sodium retention, and ultimately
low rate without an absorption phase, as the original Star- edema.11
ling equation proposed. This is an essential feature of the In severe chronic heart failure,12 the heart may have
revised paradigm and highlights the limitations of reached its maximum capacity to increase contractility in
attempting to prevent or treat edema by transfusing col- response to increasing stretch. Despite plasma volume
loids. expansion from neurohumoral activation in the face of
Edema is formed when there is either an alteration in low cardiac output, the slight increase in plasma volume
capillary dynamics favoring an increase in net filtration produces a considerable elevation in LVEDP but no in-
or defective removal of addi- crease in cardiac output.
tional filtered fluid by CLINICAL SUMMARY Hence, this leads to edema
lymphatic drainage. formation.
Increased fluid filtration is  The pathophysiology of edema has evolved with greater
In heart failure, cirrhosis,
often due to increased capil- understanding of the glycocalyx, challenging principles of and nephrotic syndrome,
lary hydrostatic pressure or the original Starling’s principle. increased release of natri-
capillary permeability (capil- uretic hormones such as
lary hydraulic permeability).  Advances in mechanisms of diuretic resistance, particularly atrial natriuretic peptide
the role of chloride, provide potential future therapeutic
In addition, it can be due to (ANP) fails to promote
targets for patients with heart failure.
disruption of the endothelial natriuresis due to renal
glycocalyx, decreased inter-  Predominance of the “overfill theory,” particularly in resistance to the action of
stitial compliance, a lower adults, in addition to small clinical trials, does not support ANP13 (see nephrotic syn-
plasma oncotic pressure, or the common practice of infusing albumin with diuretics drome).
a combination of these for the management of refractory edema.
changes (Fig 2). PATHOGENESIS OF
EDEMA IN CIRRHOSIS
PATHOGENESIS OF EDEMA IN HEART FAILURE In liver disease, the development of portal hypertension is
In heart failure, edema is due to an increase in venous pres- necessary for ascites development.14,15 In addition, pa-
sure in the heart (ventricular diastolic pressure), resulting tients with cirrhosis and portal hypertension have hemo-
in back transmission of the pressure to the venous system dynamic, functional, and biochemical abnormalities
(to the peripheral capillaries in right heart failure and pul- contributing to fluid retention.
monary capillaries in left heart failure) resulting in a paral- Cirrhosis with portal hypertension is characterized by a
lel rise in capillary hydrostatic pressure; also, there is marked reduction in systemic vascular resistance (SVR)
volume expansion due to renal sodium retention second- and mean arterial pressure plus an increase in cardiac
ary to activation of the RAAS. The site of edema accumu- output.16 The most commonly affected area is the
8
lation depends on the nature of the cardiac disease. splanchnic circulation. Increased nitric oxide production
Impairment of left ventricular function in coronary artery is most often implicated as the perpetrator for causing
disease, hypertensive heart disease, and left-sided splanchnic vasodilatation.16 Other vasodilators consid-
valvular disease results in a parallel increase in left ventric- ered have been glucagon, vasoactive intestinal peptide,
ular end-diastolic pressure (LVEDP), pulmonary venous substance P, and prostacyclins.17,18 In addition, bacterial
hydrostatic pressure, and pulmonary edema. Right ven- translocation through the gastrointestinal tract and endo-
tricular failure, via similar mechanisms, leads to periph- toxemia has been postulated to cause increased systemic
eral edema in the lower extremities and ascites. prostacyclins19 and nitric oxide.20,21 The other contrib-
In addition to the etiologies mentioned above, plasma uting factors to the decrease in SVR are the opening of por-
volume expansion increases capillary hydrostatic pres- tosystemic shunts.22
sure in chronic heart failure. The reduction in cardiac The decrease in SVR from progressive vasodilation seen in
output decreases tissue perfusion, leading to the activa- cirrhosis leads to the reduced stretch at the carotid and renal

Adv in Kidney Disease and Health 2023;30(2):110-123


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en marzo 06, 2023. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
112 Koirala et al

Table 1. Mechanism of Action of Diuretics


Oral Time to
Diuretic Typical Dose Bioavailability Half Life Onset (IV) Equipotent Doses
Loop diuretics (mechanism of action [MOA]: inhibition of NKCC2 channels)

Furosemide (IV or PO) 20-240 mg (IV) (up to q6 h) 10-100% 1.5-3 h 25 min 40 mg PO furosemide ¼ 20 mg IV
(typical max dose 600 mg) furosemide ¼ 1 mg
Bumetanide (IV or PO) 0.5-10 mg (IV) (up to q6 h) 80-100% 1-1.5 h 25 min bumetanide ¼ 20 mg torsemide
(typical max dose 15 mg)
Torsemide (PO) 10-150 mg (up to TID) 80-100% 3-6 h N/A
(typical max dose 300 mgl)

Thiazide and thiazide-like diuretics. MOA: inhibit NCC channels in the distal tubule

Metolazone (PO) 2.5-10 mg (up to BID) 270% 8-14 h 5 mg metolazone ¼ 50 mg


Chlorthalidone (PO) 50-100 mg (up to BID) 265% 40-60 h chlorthalidone ¼ 500 mg
chlorothiazide ¼ 2.5 mg
indapamide
Chlorothiazide (IV) 500-1000 mg (up to TID) 22 h
Indapamide (PO) 5-10 mg daily 293% 14-24 h

Mineralocorticoid receptor antagonists. MOA: inhibit mineralocorticoid receptors

Spironolactone (PO) 100-200 mg 290-95% 17-22 h 25 mg spironolactone ¼ 50 mg


eplerenone
Eplerenone (PO) 200-400 mg Unknown 4-6 h

Direct ENaC inhibitors. MOA: inhibit ENaC channels in the collecting duct

Amiloride (PO) 2.5-10 mg BID 30-90% 6h


Triamterene (PO) 50-100 mg BID 30-70% 2-4 h

Carbonic anhydrase inhibitor. MOA: inhibits carbonic anhydrase, indirectly decreasing sodium reabsorption in the proximal tubule

Acetezolamide (IV or PO) 500-1000 mg (up to BID) .90% 4-8 h

Sodium-glucose cotransporter 2 inhibitors. MOA: inhibit SGTL2 channels in the proximal tubule

Empagliflozin 10-25 mg daily .70% 12 h

Abbreviations: BID, twice daily; IV, intravenous; N/A, not available; NCC, sodium-chloride cotransporter; NKCC, sodium-potassium-chloride
cotransporter; PO, per os; SGLT2, sodium-glucose co-transporter 2; TID, three times daily.

baroreceptors. This results in the activation of sodium- PATHOGENESIS OF EDEMA IN NEPHROTIC


retaining neurohumoral mechanisms (RAAS, sympathetic SYNDROME
nervous system, and antidiuretic hormone) to restore In nephrotic syndrome, edema formation occurs due to 2
normal perfusion pressure.23 The downstream effects are primary mechanisms, and their contribution may vary be-
decreased renal sodium and water excretion and expansion tween patients.26–28
of extracellular volume. In a compensated state of cirrhosis,
plasma volume expansion should reduce the activity of the The Underfill Hypothesis
endogenous neurohumoral systems, thus establishing a Under this hypothesis, movement of fluid from the
new steady state with progressive normalization of sodium vascular space into the interstitium, due to a decrease in
and water excretion to match dietary intake.15 plasma colloid oncotic pressure from hypoalbuminemia,
Further decompensation of liver disease from the pro- leads to underfilling of the vasculature and activation of
gression of cirrhosis, infections, gastrointestinal bleeding, the RAAS resulting in sodium retention by the kidneys
and drugs can decrease effective intravascular volume, (Fig 3).
despite having total extracellular volume excess and Some patients with minimal change disease (MCD),
increased cardiac output. This results in ongoing activa- especially children, have elevated plasma renin activity
tion of neurohumoral systems, increased renal vasocon- and enhanced tubular sodium absorption leading to so-
striction, reduced salt and water excretion, sodium dium and water retention. These findings suggest under-
avidity, and progressive hyponatremia from increased filling the circulation due to reduced oncotic pressure
antidiuretic hormone release,24,25 which can have negative secondary to hypoalbuminemia. In 1 study with 30 chil-
prognostic implications.24,25 dren with early MCD relapse,29 21 developed edema.

Adv in Kidney Disease and Health 2023;30(2):110-123


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en marzo 06, 2023. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
Etiology and Management of Edema: A Review 113

A Classic Starling’s Principle Jv = K[(ρc-ρi)-σ(πc- πi)]

Vascular
lumen

Pc c

Pi i

Filtration rate (Jv)


Interstitium

B
Revised Starling’s Principle Jv = (ρc-ρg)-σ(πc- πg)

Vascular Vascular
Pc c lumen Pc c lumen
Intact glycocalyx damaged glycocalyx

Pg g
Small pore Pg g
Large pore network
transporting
plasma
proteins

Intercellular Intercellular
cleft cleft

Pi i Pi i

Filtration rate (Jv) Filtration rate (Jv) Interstitium


Interstitium

Figure 1. Classic versus revised Starling principle. Classic and modified views of starling’s forces along the glomerular capil-
lary wall. (A) The classic theory of continuous endothelium as a semipermeable membrane. This classic model is now consid-
ered flawed and has been superseded by the revised Starling principle. (B) The modified glycocalyx-cleft model depicted in
physiological (left panel) and pathological (right panel) states identifies glycocalyx as a semipermeable layer. Jv, fluid flux
across the capillary membranes; rc, capillary hydrostatic pressure; ri, hydrostatic pressure in the interstitium outside the
capillary; pc, capillary osmotic pressure; pi, interstitial osmotic pressure; s, capillary wall permeability. Revised model: pg, on-
cotic pressure in the sub-glycocalyx space; rg, hydrostatic pressure of the thin layer of interstitial fluid in the sub-glycocalyx
space. Arrow and line weights represent relative changes in effect and direction. Adapted from Finfer and colleagues Nat Rev
Nephrol 14, 541–557 (2018). (For interpretation of the references to color in this figure legend, the reader is referred to the Web
version of this article.)

Sixty-two percent of the children with edema had plasma volume was 66.9 6 2.5% of the control group.
biochemical evidence of volume depletion and low frac- These changes were not seen when plasma albumin
tional excretion of sodium (FENa). Manning and Guyton30 decreased 33% during a 5-day plasmapheresis period.
performed plasmapheresis followed by isotonic fluid infu- Vande Walle and colleagues31 reported that in 6 children
sion to reduce plasma protein and noticed that when albu- with nephrotic syndrome (non-MCD nephrotic syndrome
min decreased 68% over a 12-day plasmapheresis period with hypovolemic symptoms [4 with congenital nephrotic
(mean albumin 2.4 g/dL), mean arterial pressure decreased syndrome of the Finnish type]), FENa was 0.2 6 0.2%.
by 26 mm Hg, plasma renin activity increased 11-fold, and Lithium clearance and maximal water excretion were

Adv in Kidney Disease and Health 2023;30(2):110-123


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en marzo 06, 2023. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
114 Koirala et al

VASOCONSTRICTION
NFP= Lp S (Pgc-Pbs)- p− bs
(hydraulic pressure) - (oncotic pressure)
+++ NE, NSAIDs
Endothelin +13 mm Hg
++ CNIs, ATP
+ Ang II

VASODILATION
--- ANP, Nitric Oxide
Dopamine
- ARB/ACEi -3 mm Hg
10
46
onc
23 BSonc
gc onc <1
35 0 Pbs
onc
20
35
45
VASOCONSTRICTION 10 onc
+++ Ang II, NE 6
++ CNIs, ANP
8
+ Endothelin

VASODILATION onc onc


--- ARB/ACEi, 37 25
Dopamine
- Nitric Oxide

20 15

Figure 2. Revised Starling principle. Starling forces (revised view) govern the passive exchange of water between the capil-
lary microcirculation, glycocalyx, and the interstitial fluid by determining the net directionality and rate of water exchange. s,
protein reflection coefficient ¼ 1; pðoncÞbs ¼ 0; pgc, oncotic pressure in the sub-glycocalyx space; rgc, hydrostatic pressure of
the thin layer of interstitial fluid in the sub-glycocalyx space; ACEi, angiotensin-converting enzyme inhibitors; Ang II, angio-
tensin II; ANP, atrial natriuretic peptide; ARB, angiotensin receptor blockers; CNIs, calcineurin inhibitors; Lp, porosity of the
capillary wall; NE, norepinephrine; NFP, net filtration pressure; NSAIDs, nonsteroidal anti-inflammatory drugs; S, surface area
¼ (100x higher than other beds).

suppressed, and plasma aldosterone, renin, and norepi- ANPs are elevated in nephrotic syndrome, but experi-
nephrine levels were elevated. Among children with mental and human data demonstrate markedly blunted
MCD, 12 presented with hypovolemic symptoms and natriuretic and diuretic responses to systemic infusion of
strong sodium retention (FENa 0.3%), whereas 15 were sta- ANP in the nephrotic syndrome. The altered response to
ble (FENa 1.1%). In the children with non-MCD lesions, the ANP is also related to a defect in the number and affinity
plasma colloid osmotic pressure was significantly lower in of receptor-binding sites for the peptide and a possible
the hypovolemic types than in those with stable edema (4.2 defect at the intracellular cyclic guanosine monophos-
vs 13.0 mm Hg; P , 0.05); in MCD, no such difference ex- phate (cGMP) level, the second messenger of ANP.26 In
isted (8.1 mm Hg and 9.9 mm Hg, respectively). addition, the gene encoding for a cyclophilin-like protein,
which is increased in sodium-retaining conditions, is upre-
gulated in the kidneys of nephrotic rats, and the infusion of
The Overfill Hypothesis ANP further increases cyclophilin-like protein mRNA.33
Primary sodium retention by the kidney due to increased Valentine and colleagues34 revealed a blunted natri-
epithelial sodium channel (ENaC) activity and renal resis- uretic response to volume expansion with isotonic saline
tance to systemic ANP results in plasma volume expan- in proteinuric rats in a rat model of Heymann nephritis.
sion (Figs 4 and 5). Plasma ANP concentration was increased to the same
Ichikawa and colleagues32 reported a unilateral model extent in both normal and nephrotic rats compared to hy-
of polyarteritis nodosa (PAN)-induced nephrotic syn- povolemic controls. However, accumulation of cGMP by
drome in Munich-Wistar rats. Glomerular filtration rate isolated glomeruli and inner medullary collecting duct
(GFR) and single-nephron GFR were reduced selectively cells from nephrotic rats after incubation with ANP was
in PAN-perfused kidneys with nephrotic syndrome by significantly reduced compared with normal rats. This
30%. Micropuncture studies revealed decreased sodium difference was not related to differences in either density
filtration in the nephrotic kidney and less reabsorption or affinity of renal ANP receptors but was abolished
in the proximal convoluted tubule due to reduced oncotic when the accumulation of cGMP was measured in the
pressure in the peritubular capillaries and loop of Henle. presence of inhibitors of cyclic nucleotide phosphodies-
However, the quantity of sodium remaining in the terases. Infusion of the phosphodiesterase inhibitor zapri-
tubular lumen at the end of the late distal tubules re- nast into 1 renal artery in nephrotic rats normalized both
mained the same in both PAN-perfused and control kid- the natriuretic response to volume expansion and the in-
neys, implying increased sodium reabsorption in the crease in urinary cyclic guanosine monophosphate excre-
distal tubules. tion from the infused, but not the contralateral, kidney.

Adv in Kidney Disease and Health 2023;30(2):110-123


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en marzo 06, 2023. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
Etiology and Management of Edema: A Review 115

Increased activity of ENaCs has been noted in nephrotic Several studies have shown that concomitant use of
syndrome (Fig 5).35,36 One of the serine proteases filtered intravenous (IV) hypertonic saline solution with high
in patients with nephrotic syndrome, plasminogen, gets doses of loop diuretics for the management of persistent
activated to plasmin by a tubular urokinase-type plasmin- congestion improves diuresis and clinical outcomes.47,51
ogen activator (uPA). Plasmin has been known to cause The exact mechanisms for the benefit of hypertonic saline
proteolytic cleavage of the gamma chain of ENaCs, lead- remain elusive. It was initially hypothesized that mobiliza-
ing to its activation and sodium retention. tion of fluid from the interstitial space to the intravascular
Staehr and colleagues37 administered amiloride, which compartment through osmotic forces would restore effec-
inhibits the ENaCs and uPA activity. Amiloride reduced tive intravascular volume, with resultant enhanced renal
urine uPA activity, plasminogen activation, protease activ- blood flow (RBF) and delivery of diuretic agents to the
ity, and sodium retention in PAN rats, while proteinuria loop of Henle.52,53 However, this hypothesis was chal-
was not altered. In paired urine samples,37 uPA protein lenged by a study involving slow infusion of hypertonic
level was significantly elevated in urine from children saline and minimal osmotic improvement but with similar
with active nephrotic syndrome compared with the remis- beneficial results.54 Therefore, a definitive mechanism ex-
sion phase. In addition, in 6 adult nephrotic patients, urine plaining the reported benefits of hypertonic saline in
uPA protein and activity correlated positively with decongestive therapy remains unclear and needs to be
24-hour urine protein excretion. further evaluated.

MANAGEMENT OF EDEMA ROLE OF CHLORIDE


Diuretics are ubiquitous in the management of congestion Chronic diuretic use may lead to chloride depletion via
and edema, but the response is often inadequate.38–40 chloriuresis, which in turn plays an often overlooked
Diuretic resistance (DR), commonly defined qualitatively role in DR.55 Chloride (Cl2) signals the macula densa to
as a decreased natriuretic response necessitating respond to volume overload via tubuloglomerular feed-
progressive increases in diuretic dosing and/or variations back, thus mediating renin release.56 As such, chloride
in medication combination, is a major clinical challenge levels are strongly linked to diuretic responsiveness,
and often portends a poor prognosis.41 At this time, there with chloride-containing solutions in turn increasing renal
is no standardization of diuretic therapy following identi- NaCl excretion.57–59 Prior animal studies explored this
fication of DR. This review covers the etiology of DR from phenomenon by directly infusing hypertonic or
a physiological standpoint, focusing on novel hypotonic NaCl into the renal artery while maintaining
mechanisms. plasma Na1 and Cl2 levels via hemodialysis.60 Results
showed that increases in renal plasma sodium and chlo-
DIURETIC BRAKING ride [P[NaCl]] led to increased sodium excretion despite re-
DR mechanisms have been coalesced under the term ductions in eGFR, RBF, and intrarenal RAAS
“diuretic braking,” a descriptive term for the progressive activation.60,61 While hypertonic saline or ammonium
decrease in response to the same dose of diuretic chloride reduced RBF and GFR and increased urine so-
throughout therapy.42 Diuretic braking is beneficial as it al- dium volume (UNaV), intrarenal infusion of sodium ace-
lows a wider therapeutic window but, at the same time, tate actually increased RBF and GFR and reduced
can result in impaired decongestion. The term diuretic UNaV.48,59,61 These results relate renal vasoconstriction
braking in clinical practice fails to recognize a specific and natriuresis to increased Cl2, independent of Na1 or
mechanism of resistance; however, one explanation for plasma osmolality. Tubular fluid Cl2 plays a dominant
this phenomenon could be enhanced distal sodium chlo- role in the reduction of eGFR and RBF via the tubuloglo-
ride (NaCl) reabsorption.43 This is often in the setting of merular feedback mechanism and increases in afferent
intermittent diuretic dosing.44 Diuretic dose and fre- arteriolar resistance.62 The effect of increased Cl2 in
quency are interdependent in DR, and both must be increasing UNaV was independent of renal hemody-
considered.45 Herein, we further discuss the roles of so- namics. This natriuresis may be partially explained by
dium and chloride in resistance to diuretic therapy. Mech- chloride-induced metabolic acidosis, resulting in
anisms of diuretic braking are displayed in Figure 6. respiratory compensation and resultant decrease in
PCO2, which in turn reduces proximal tubule NaCl and
SODIUM AVIDITY AND SALT INTAKE fluid reabsorption.63,64
Sodium has been understood to play a paramount role in Thus, an increase in Cl2, independent of sodium or os-
fluid homeostasis in the setting of congestion.46,47 The molarity, has a potent effect to induce renal vasoconstric-
importance of kidney sodium avidity in determining tion, reduce intrarenal angiotensin II, and increase renal
diuretic response has been highlighted by recent studies NaCl and fluid excretion. Indeed, acute hyperchloremia
reporting that healthy individuals on a very-low-salt diet in human subjects reduces RBF and renal renin and angio-
had a noticeable decrease in response to loop diuretics.48 tensin release.65 Moreover, hypochloremic metabolic alka-
Enhanced sodium avidity may also explain excessive so- losis during loop diuretic administration reduces the
dium retention in the absence of diuretics.49 As such, var- diuretic response by up to 40%.66 This may be because de-
iations in diuretic responsiveness, despite a similar livery of Cl2 to the loop of Henle determines the activity of
estimated GFR (eGFR), may be a result of differences in the Na1/2Cl2/K1 (NKCC2) cotransporter, which is the site
basal sodium avidity.45,50 of action of loop diuretics.66,67 In contrast, depletion of

Adv in Kidney Disease and Health 2023;30(2):110-123


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en marzo 06, 2023. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
116 Koirala et al

Underfill Theory of Edema

↓ Plasma blood and Plasma Restoration of Blood &


Kidney Damage
volume Plasma Volume
↓ Blood Pressure ↑ ECF
Proteinuria Orthostatic Hypotension Normal Blood Pressure

Normal levels
Hypoalbuminemia ↑Renin ↑ Aldo
↓ ANP Renin Aldo
↑ NE ↑ ADH
NE ADH ANP

↓ Oncotic Pressure
↓ GFR, & ↓ FF
↑ Sodium reabsorption in
Shift of PCT & DCT
intravascular fluid ↑ Water reabsorption
into Interstitium
Hyponatremia

EDEMA FORMATION

Figure 3. Underfill theory of edema. Proteinuria, depleted intravascular volume, and decreased blood pressure can activate
the renin-angiotensin-aldosterone (Aldo) system, increase anti-diuretic hormone (ADH) and norepinephrine (NE) secretion,
decrease norepinephrine (NE) and atrial natriuretic peptide (ANP). These changes decrease glomerular filtration rate (GFR)
and filtration fraction (FF) and increase sodium reabsorption in the proximal and distal convoluted tubules (PCT and DCT).
This restores blood, plasma, and extracellular fluid (ECF) volume. (For interpretation of the references to color in this figure
legend, the reader is referred to the Web version of this article.)

intracellular Cl2 activates NKCC2 via effects in WNK3, a serum sodium, was strongly associated with impaired
chloride-sensitive kinase.66,67 decongestion and increased mortality.55,68 Moreover, hy-
Chloride has also been shown to reduce renin release in pochloremia was associated with poor diuretic response
contrast to non–chloride-containing sodium salts, which in patients with HF while its correction, independent of
do not affect renin levels.59 Previous studies of patients serum sodium with lysine chloride, improved clinical
admitted with acute decompensated heart failure responses to diuretics.69 Therefore, repletion of chloride
(ADHF) have shown that low serum chloride, but not may be considered in patients with DR.

ANP

2 ANP (Atrial Natriuretic


• Increased afferent and 1 peptide) Resistance
efferent arteriolar
resistance due to
increased sympathetic ↑ open channel
activity probability of ENaC
• Increased sympathetic (Epithelial Sodium Channel)
activityÆ activation of 3 4
tubular adrenergic
receptorsÆ Sodium and
water reabsorption
throughout the nephron
↑ Number and activity
of Na-K-ATPase

Figure 4. Overfill theory of edema. Mechanisms supporting overfill theory of edema. 1. Increased sympathetic activity leads
to arteriolar resistance, activation of adrenergic receptors, and increased sodium and water reabsorption; 2. resistance to
atrial natriuretic peptide (ANP) and decreased sodium excretion; 3. increased probability of ENaC opening and increased
number and activity of sodium potassium ATPase (Na-K-ATPase). Aldo, aldosterone; ECF, extracellular fluid; ENaC, epithelial
sodium channel; FF, filtration fraction; GFR, glomerular filtration rate; NE, norepinephrine; PCT and DCT, proximal and distal
convoluted tubule. (For interpretation of the references to color in this figure legend, the reader is referred to the Web version
of this article.)

Adv in Kidney Disease and Health 2023;30(2):110-123


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en marzo 06, 2023. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
Etiology and Management of Edema: A Review 117

Overfill Theory of Edema


Kidney Damage Lumen Cortical Collecting Duct Blood
Principal Cell
Proteinuria

Plasminogen excretion
& conversion to Plasmin Plasminogen

Urokinase-type plasminogen
activator
Inhibition of domain Plasmin
by Plasmin Closed ENaC due to the
presence of the domain

ENaC (Epithelial
Sodium Channel) O
Open ENaC
activation

Sodium retention &


edema

Figure 5. Overfill theory of edema. Mechanism of epithelial sodium channel (ENaC) opening. Proteinuria and plasminogen
excretion increases the concentration of plasminogen in the lumen, which converts to plasmin. Plasmin inhibits a part of
the gamma (gÞ domain of the ENaC channel that has an inhibitory effect on the channel, which leads to persistent activation
of ENaC and sodium retention. (For interpretation of the references to color in this figure legend, the reader is referred to the
Web version of this article.)

A family of serine-threonine kinases (with no lysine WNK kinases play a central role in intracellular signaling
[WNK]) have been shown to play a key role in regulation and regulation of ion transport and neurohormonal
of electrolyte homeostasis and the transporters where loop pathways in response to hypochloremia73 as they regulate
and thiazide diuretics function.70,71 Hypochloremia is the targets of both loop and thiazide diuretics.74
sensed by specific WNKs that regulate the sodium- It has been shown that reduced chloride levels activate the
potassium-chloride cotransporter and thereby modify WNK1 kinase through autophosphorylation. Once acti-
salt and water retention.72 vated, WNK1 kinase activates and upregulates NKCC2

Diuretic Resistance Mechanisms

Prolonged use of loop or Thiazide diuretics

Low Plasma Potassium Low Plasma Chloride

Autophosphorylation & activation Decreased Cl delivery to Metabolic Alkalosis


of WNK Kinases Macula Densa
Increased delivery of bicarbonate
to NKCC2

Upregulation of Upregulation of Increased Renin, Angiotensin II


NKCC2 in TAL NCC in DCT & Aldosterone Activation of NDBCE in DCT

Increased Electrolyte & Fluid Reabsorption

Decreased Effect of Diuretic Therapy

Figure 6. Diuretic resistance mechanisms. Multiple mechanisms of diuretic resistance in patients with edema. DCT, distal
convoluted tubule; NCC, sodium-chloride cotransporter; NDBCE, sodium-dependent bicarbonate-chloride exchanger;
NKCC, sodium-potassium-chloride cotransporter; TAL, thick ascending loop of Henle. (For interpretation of the references
to color in this figure legend, the reader is referred to the Web version of this article.)

Adv in Kidney Disease and Health 2023;30(2):110-123


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en marzo 06, 2023. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
118 Koirala et al

(Continued )
Mechanism of Edema
Causes of Edema Formation Mechanism of Edema
Causes of Edema Formation
Heart failure Decreased EF and forward
flow results in decreased  Wet beriberi  Thiamine deficiency
effective intravascular leading to high-output
volume and activation of heart failure
SNS, RAAS and ADH Refeeding edema Patients who have fasted for
leading to sodium and some time when fed a
water retention. Increased carbohydrate-rich diet
LVEDP leads to pulmonary retain sodium in response
venous congestion. to insulin-induced sodium
Increased right-sided reabsorption in the
filling pressures contribute proximal convoluted
to hepatic congestion, tubule.
ascites, and peripheral Lymphatic obstruction Impaired lymphatic drainage
edema. of interstitial fluid back to
Nephrotic syndrome Overfill hypothesis: primary the systemic circulation
renal sodium retention by (eg, radical lymph node
activation of RAAS and dissection for breast
ENaC activation resulting cancer).
in volume expansion and Drug-induced edema
edema. NSAIDs Depletion of vasodilatory
Underfill hypothesis: prostaglandins leads to
Underfilling of the increased renal sodium
circulation by a decreased retention. NSAIDs
plasma oncotic pressure exacerbate volume
leads to secondary renal salt overload in heart failure
and water retention. and cirrhosis and can
Chronic kidney disease Decreased GFR results in salt cause diuretic resistance.
and water retention Antihypertensive agents
Cirrhosis Portal hypertension leads to  Direct arterial/  Arteriolar vasodilatation
ascites. Decreased arteriolar leads to increased
effective intravascular vasodilators: capillary hydrostatic
volume results in the hydralazine, pressure and increased
activation of SNS, RAAS clonidine, filtration at the capillary
and ADH leading to methyldopa level. Direct vasodilators
sodium and water guanethidine, also lead to a drop in
retention. minoxidil systemic blood pressure,
Localized edema RAAS activation, and renal
 Venous obstruction  Increased venous sodium retention.
(DVT or venous stasis) hydrostatic pressure  Adrenergic
leading to increased antagonists
ultrafiltrate formation.  Calcium channel  CCB-induced edema is due
 Allergic reactions  Loss of vascular integrity, blockers (CCB) to arteriolar vasodilatation
(laryngeal edema) allowing fluid to move into leading to increased
the interstitial tissues due formation of capillary
to the presence of ultrafiltrate. It is more
inflammatory mediators common with
(due to mast cell-mediated dihydropyridine than with
angioedema, bradykinin- nondihydropyridines. CCB
mediated angioedema, and RAAS blockers
and from unknown improve edema by
mechanisms). causing capillary
Nutritional deficiencies venodilatation.
 Kwashiorkor  Increased generation of Thiazolidinediones Edema formation by an
cysteinyl leukotrienes that unknown mechanism.
increase capillary Contraindicated in NYHA
permeability and class 3 and 4 heart failure.
hypoalbuminemia, DPP4 inhibitors Angioedema either alone or
leading to decreased in combination with RAAS
plasma colloid oncotic inhibitors due to reduced
pressure. clearance of bradykinin by
(Continued ) DPP4 inhibitors.
(Continued )

Adv in Kidney Disease and Health 2023;30(2):110-123


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en marzo 06, 2023. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
Etiology and Management of Edema: A Review 119

(Continued ) A reduction in chloride delivery to the loop of Henle


Mechanism of Edema (such as during volume depletion) increases absorption
Causes of Edema Formation of NaCl via upregulation of NKCC2 and NCCs, whereas
low chloride delivery to the macula densa stimulates
Steroid hormones Primary renal salt and water renin secretion from the juxtaglomerular apparatus.67,68
 Glucocorticoids retention through the
Hyperactivity of the RAAS has long been known to
 Anabolic steroids mineralocorticoid receptor
 Estrogens and activation of ENaC
portend an unfavorable impact on ADHF and is a vital
 Progestins channels in principal cells. treatment target in these patients.69 The ROSE-AHF
Cyclosporine Renal vasoconstriction and (Renal Optimization Strategies Evaluation in Acute Heart
stimulation of thiazide- Failure) trial reported that although baseline chloride
sensitive NaCl channels levels did not correlate with worsening HF, hypochlore-
lead to salt and water mia had a negative association with response to diuretics
retention. and diuretic efficiency, increased mortality, and rehospi-
Growth hormone Primary renal salt and water talization rate.80
retention by growth The chloride-bicarbonate exchanger pendrin, located on
hormone and through IGF-
ß-intercalated cells in the kidney cortical collecting duct
1 by activation of ENaC
channels in principal cells.
and connecting tubules, which mediates the secretion of
Immunotherapies bicarbonate and reabsorption of chloride and plays an
 Interleukin 2  Increased capillary important role in vascular homeostasis, has been sug-
permeability from high- gested as a target for diuretic therapy.81,82 Pendrin elimi-
dose IL-2. nates excess bicarbonate during alkalosis and is a novel
 OKT3 monoclonal  Depletion of T cells by therapeutic target for diuretics.81,82 Chloride can be
antibody OKT3 leads to paradoxical directly reabsorbed in exchange for bicarbonate by pen-
(Muromonab-CD3) T-cell activation resulting drin, or chloride absorption can be coupled to sodium ab-
in cytokine release sorption by driving the activity of another transporter, the
syndrome and increased
Na1-dependent bicarbonate-chloride exchanger, replac-
capillary permeability.
ing one intracellular chloride for one extracellular bicar-
Abbreviations: ADH, anti-diuretic hormone; DPP4, Dipeptidyl pepti- bonate plus one sodium. This results in the net
dase 4; DVT, deep venous thrombosis; EF, ejection fraction; ENaC, absorption of NaCl.83
epithelial sodium channels; GFR, glomerular filtration rate; IGF-1, in-
sulin-like growth factor 1; LVEDP, left ventricular end-diastolic pres-
Administration of a selective pendrin inhibitor as mono-
sure; NSAIDs, nonsteroidal anti-inflammatory drugs; NYHA, New therapy to rodents results in no change in electrolyte
York Heart Association; OKT3, Muromonab-CD3; RAAS, renin- handling or urine output unless the rat has been chroni-
angiotensin-aldosterone system; SNS, sympathetic nervous sys- cally treated with furosemide, which causes increases in
tem. urine output.84 As such, pendrin is an excellent candidate
for human DR in that it offers a salvage pathway when
and the NaCl cotransporter (NCC) in the thick ascending distal delivery of sodium is increased.
limb and distal convoluted tubule, respectively, which are
the primary targets of loop and thiazide diuretics, thereby Renal Venous Congestion
enhancing renal electrolyte and fluid reabsorption. The A small increase in renal interstitial pressure, such as that
numbers of NKCC2 and NCC receptors are thought to following renal lymphatic ligation, enhances renal sodium
have an inverse relationship with diuretic efficiency. Activa- excretion.85 In addition, a modest increase in renal venous
tion of chloride-sensing WNKs by hypochloremia with pressure that increases the renal interstitial pressure in-
initiation of the downstream signaling cascade and upregu- duces a brisk natriuresis.86 However, steeper increases in
lation of NKCC2 and NCC may impair diuretic response renal venous pressure can collapse capillaries and tubules
and inhibit the treatment of congestion.67,75 in the encapsulated kidney, thereby reducing RBF, GFR,
Reports of WNK4’s effect over NCC activity were and the excretion of fluid and sodium.87 A fall in GFR dur-
initially discordant, as evidence showed inhibitory modu- ing diuretic therapy is generally attributed to volume
lation in vitro,71,76 while in vivo evidence demonstrated depletion88; however, an abrupt and considerable increase
WNK4 as an activator of NCC.77 Later, it was explained in intrarenal pressure could be accompanied by a fall in
that this discrepancy could be supported by WNK4 regu- RBF and GFR.89 Because the main intranephron hydro-
lation by chloride. One study showed that while WNK4 static resistance to flow is downstream from the loop of
coexpression does not upregulate NCC in basal condi- Henle,90 inhibition of fluid reabsorption within the loop
tions, decreasing plasma chloride promotes WNK4s acti- of Henle will considerably increase intratubular and renal
vating phosphorylation and WNK4-mediated NCC interstitial fluid pressure. Indeed, furosemide causes a
activation.78 Mutation of the WNK4 chloride binding site sharp rise in the volume of the distal, but not the proximal,
changed it into an active kinase that upregulated NCC ac- tubules in the rat,91 likely because it reduces fluid reab-
tivity. These series of trials not only assisted understanding sorption in the ascending limb of the loop of Henle, the
the effects of WNK4 over NCC function but also estab- thin loops of Henle, and the collecting ducts that are in
lished WNK kinases as key chloride-sensing proteins either side of the loop of Henle, which are the major
that regulate the activity of the transporters.79 nephron segments contributing to intratubular fluid

Adv in Kidney Disease and Health 2023;30(2):110-123


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en marzo 06, 2023. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
120 Koirala et al

resistance to flow.92 Moreover, furosemide activates renal difference in natriuresis or urine volume at 24 hours.101 Al-
afferent nerves likely by activation of renal baroreceptors bumin administration has been insufficiently studied in pa-
and chemoreceptors.93 Interestingly, the falls in RBF and tients with a serum albumin concentration less than 2.0 g/
GFR with IV furosemide are dependent on renal afferent, dL; therefore, it remains possible that a combination of
but not efferent, nerves.94 Therefore, acute renal conges- loop diuretics with albumin remains a viable option for
tion accompanying IV loop diuretics may contribute to those with severe hypoalbuminemia. One such study as-
adverse renal hemodynamics and DR via direct effects of sessed the efficacy of furosemide with or without albumin
the rise in intrarenal pressure and via afferent renal neural in patients with severe hypoalbuminemia (mean serum al-
mechanisms rather than by volume depletion. Indeed, the bumin 1.7 g/dL) and acute lung injury. The authors reported
acute falls in RBF and GFR with IV furosemide persist un- a significant improvement in cumulative fluid balance in
modified if the NaCl and fluid excreted is replaced quan- the treatment group; however, the control group received
titatively by a matching saline infusion.94 Similarly, the significantly more fluid boluses, likely explaining the
sharp fall in Na1 and fluid excretion that occurs after the difference.102 Thus, coadministration of albumin and loop
loop diuretic effect has abated and is considered to be diuretics for the treatment of refractory edema in patients
“compensatory” in healthy human volunteers in whom with hypoalbuminemia is not currently supported by pub-
all salt and water loss is prevented by a saline infusion.95 lished literature. An exception to this may be in patients
In ADHF, postdiuretic sodium and fluid retention is not with MCD, particularly in children. Concurrent administra-
compensatory for diuretic-induced salt loss, with basal so- tion of albumin with furosemide may result in transient im-
dium avidity being a stronger driver of spontaneous natri- provements in weight loss although at the expense of
uresis.96 Thus, renal congestion can be both a consequence possible worsening hypertension and respiratory failure.103
of volume overload and a consequence of the use of IV A proposed management strategy is to utilize a FENa to
loop diuretics for its treatment. In both settings, the identify patients who were volume-contracted (supported
congestion is associated with impaired renal hemody- by the underfill theory) with those who were volume-
namics and renal sodium and fluid retention. expanded (supported by the overfill theory). Administra-
tion of albumin with furosemide to the volume-contracted
ALBUMIN FOR REFRACTORY EDEMA patients, defined as a FENa , 0.2%, led to an improvement
The cornerstone of therapy for edema, other than reversing in biochemical parameters and no difference in hospital
the underlying disorder when possible, involves achieving length of stay or weight loss compared to furosemide alone
net negative sodium balance,97 typically with the initiation for patients identified as volume-expanded.104
of a loop diuretic. Loop diuretics are highly protein-bound
and, therefore, have a low volume of distribution; however,
in hypoalbuminemic patients, the volume of distribution CONCLUSION
can increase substantially. This concept provides a potential Despite recent advancements in the management of
mechanism for diuretic hyporesponsiveness in this popula- congestion and volume overload, there remains a gap in
tion and suggests that albumin infusion, concurrent with our understanding of diuretic therapy and how it may
loop diuretic administration, may be an attractive therapeu- be optimized in the setting of chronic disease. Mechanisms
tic intervention by improving drug delivery to the kidney. for DR should be more thoroughly investigated, and po-
This concept was demonstrated in analbuminemic rats tential therapeutic targets identified. Moreover, modifica-
who had a 10-fold higher volume of distribution of furose- tion of the existing therapy to match discoveries in
mide than normal animals. This resulted in insufficient con- electrolyte pathophysiology is integral to provide effective
centration of diuretics at secretory sites in the proximal diuresis and improve outcomes.
tubule and little drug secreted in the urine, hence a negli-
gible diuretic response, which was restored after infusion
REFERENCES
of an albumin/furosemide mixture.98 Unfortunately, this 1. Friis-Hansen B. Body water compartments in children: changes
has not consistently translated to human studies. A small during growth and related changes in body composition. Pediatrics.
study of 13 patients with biopsy-proven cirrhosis demon- 1961;28:169-181.
strated no difference in furosemide pharmacokinetics, urine 2. Verbalis JG. Disorders of body water homeostasis. Best Pract Res
furosemide excretion, or urine sodium excretion with albu- Clin Endocrinol Metab. 2003;17(4):471-503.
min administration (either premixed with furosemide 3. Harrison’s Principles of Internal Medicine, 20e https://
ex vivo or infused simultaneously) versus furosemide accessmedicine.mhmedical.com/book.aspx?bookID¼2129. Ac-
alone.99 A study of 9 patients with nephrotic syndrome cessed April 10, 2022.
demonstrated a modest increase in sodium excretion and 4. Taylor AE. Capillary fluid filtration. Starling forces and lymph
flow. Circ Res. 1981;49(3):557-575.
urine volume with coadministration of furosemide plus al-
5. J.E. Hall and M.E. Hall, Guyton and Hall Textbook of Medical Physi-
bumin, compared to furosemide alone, in the absence of dif- ology, 2020, Elsevier https://www.elsevier.com/books/guyton-an
ferences in urinary furosemide excretion; however, the d-hall-textbook-of-medical-physiology/hall/978-0-323-59712-8. Ac-
increased natriuresis can be explained by desalination of cessed January 18, 2023.
the sodium content of the infused colloid.100 A meta- 6. Levick JR, Michel CC. Microvascular fluid exchange and the
analysis of 10 studies (including 8 trials with a cross-over revised Starling principle. Cardiovasc Res. 2010;87(2):198-210.
design) found a modest increase in urinary sodium excre- 7. Woodcock TE, Woodcock TM. Revised Starling equation and the
tion at 8 hours (15 mmol sodium) although there were no glycocalyx model of transvascular fluid exchange: an improved

Adv in Kidney Disease and Health 2023;30(2):110-123


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en marzo 06, 2023. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
Etiology and Management of Edema: A Review 121

paradigm for prescribing intravenous fluid therapy. Br J Anaesth. 29. Vande Walle JG, Donckerwolcke RA, van Isselt JW, Derkx FH,
2012;108(3):384-394. Joles JA, Koomans HA. Volume regulation in children with early
8. Blankfield RP, Finkelhor RS, Alexander JJ, et al. Etiology and diag- relapse of minimal-change nephrosis with or without hypovolae-
nosis of bilateral leg edema in primary care. Am J Med. mic symptoms. Lancet. 1995;346(8968):148-152.
1998;105(3):192-197. 30. Manning RD Jr, Guyton AC. Effects of hypoproteinemia on fluid
9. Dzau VJ, Colucci WS, Hollenberg NK, Williams GH. Relation of volumes and arterial pressure. Am J Physiol. 1983;245(2):H284-
the renin-angiotensin-aldosterone system to clinical state in conges- H293.
tive heart failure. Circulation. 1981;63(3):645-651. 31. Vande Walle JG, Donckerwolcke RA, Koomans HA. Pathophysi-
10. Braunwald E, Plauth WH Jr, Morrow AG. A method for the detec- ology of edema formation in children with nephrotic syndrome
tion and quantification of impaired sodium excretion. Results of an not due to minimal change disease. J Am Soc Nephrol.
oral sodium tolerance test in normal subjects and in patients with 1999;10(2):323-331.
heart disease. Circulation. 1965;32:223-231. 32. Ichikawa I, Rennke HG, Hoyer JR, et al. Role for intrarenal mech-
11. Reddy HK, Weber KT, Janicki JS, McElroy PA. Hemodynamic, anisms in the impaired salt excretion of experimental nephrotic
ventilatory and metabolic effects of light isometric exercise in pa- syndrome. J Clin Invest. 1983;71(1):91-103.
tients with chronic heart failure. J Am Coll Cardiol. 1988;12(2):353- 33. Orisio S, Perico N, Benatti L, Longaretti L, Amuchastegui S,
358. Remuzzi G. Renal cyclophilin-like protein gene expression paral-
12. Schwinger RH, B€ ohm M, Koch A, et al. The failing human heart is lels changes in sodium excretion in experimental nephrosis and
unable to use the Frank-Starling mechanism. Circ Res. is positively modulated by atrial natriuretic peptide. J Am Soc Neph-
1994;74(5):959-969. rol. 1993;3(10):1710-1716.
13. Lee EY, Humphreys MH. Phosphodiesterase activity as a mediator 34. Valentin JP, Ying WZ, Sechi LA, et al. Phosphodiesterase inhibitors
of renal resistance to ANP in pathological salt retention. Am J Phys- correct resistance to natriuretic peptides in rats with Heymann
iol. 1996;271(1 Pt 2):F3-F6.E.Y. Nephritis. J Am Soc Nephrol. 1996;7(4):582-593.
14. Sola E, Gines P. Renal and circulatory dysfunction in cirrhosis: cur- 35. Svenningsen P, Bistrup C, Friis UG, et al. Plasmin in nephrotic
rent management and future perspectives. J Hepatol. urine activates the epithelial sodium channel. J Am Soc Nephrol.
2010;53(6):1135-1145. 2009;20(2):299-310.
15. Gines P, Fern andez-Esparrach G, Arroyo V, Rodes J. Pathogenesis 36. Passero CJ, Mueller GM, Rondon-Berrios H, Tofovic SP,
of ascites in cirrhosis. Semin Liver Dis. 1997;17(3):175-189. Hughey RP, Kleyman TR. Plasmin activates epithelial Na1 chan-
16. Iwakiri Y, Groszmann RJ. Vascular endothelial dysfunction in nels by cleaving the gamma subunit. J Biol Chem.
cirrhosis. J Hepatol. 2007;46(5):927-934. 2008;283(52):36586-36591.
17. Gines P, Arroyo V, Rodes J. Pathophysiology, complications, and 37. Stæhr M, Buhl KB, Andersen RF, et al. Aberrant glomerular filtra-
treatment of ascites. Clin Liver Dis. 1997;1(1):129-155. tion of urokinase-type plasminogen activator in nephrotic syn-
18. Guarner F, Guarner C, Prieto J, et al. Increased synthesis of sys- drome leads to amiloride-sensitive plasminogen activation in
temic prostacyclin in cirrhotic patients. Gastroenterology. urine. Am J Physiol Renal Physiol. 2015;309(3):F235-F241.
1986;90(3):687-694. 38. Casu G, Merella P. Diuretic Therapy in Heart Failure - Current Ap-
19. Guarner C, Soriano G, Such J, et al. Systemic prostacyclin in proaches. Eur Cardiol. 2015;10(1):42-47.
cirrhotic patients. Relationship with portal hypertension and 39. Mullens W, Damman K, Harjola VP, et al. The use of diuretics in
changes after intestinal decontamination. Gastroenterology. heart failure with congestion - a position statement from the Heart
1992;102(1):303-309. Failure Association of the European Society of Cardiology. Eur J
20. Frances R, Mu~ noz C, Zapater P, et al. Bacterial DNA activates cell Heart Fail. 2019;21(2):137-155.
mediated immune response and nitric oxide overproduction in 40. Liszkowski M, Nohria A. Rubbing salt into wounds: hypertonic sa-
peritoneal macrophages from patients with cirrhosis and ascites. line to assist with volume removal in heart failure. Curr Heart Fail
Gut. 2004;53(6):860-864. Rep. 2010;7(3):134-139.
21. Guarner C, Soriano G, Tomas A, et al. Increased serum nitrite and 41. Blankstein R, Bakris GL. Renal hemodynamic changes in heart fail-
nitrate levels in patients with cirrhosis: relationship to endotoxe- ure. Heart Fail Clin. 2008;4(4):411-423.
mia. Hepatology. 1993;18(5):1139-1143. 42. Shah S, Pitt B, Brater DC, et al. Sodium and fluid excretion with tor-
22. Vorobioff J, Bredfeldt JE, Groszmann RJ. Increased blood flow semide in healthy subjects is limited by the short duration of
through the portal system in cirrhotic rats. Gastroenterology. diuretic action. J Am Heart Assoc. 2017;6(10):e006135.
1984;87(5):1120-1126. 43. Yancy CW, Jessup M, Bozkurt B, et al. 2013 ACCF/AHA guideline
23. Henriksen JH, Bendtsen F, Gerbes AL, Christensen NJ, Ring- for the management of heart failure: executive summary: a report
Larsen H, Sørensen TI. Estimated central blood volume in cirrhosis: of the American College of Cardiology Foundation/American
relationship to sympathetic nervous activity, beta-adrenergic Heart Association Task Force on practice guidelines. Circulation.
blockade and atrial natriuretic factor. Hepatology. 1992;16(5):1163- 2013;128(16):1810-1852.
1170. 44. Ellison DH, Felker GM. Diuretic treatment in heart failure. N Engl J
24. Wong F, Liu P, Tobe S, Morali G, Blendis L. Central blood volume Med. 2018;378(7):684-685.
in cirrhosis: measurement with radionuclide angiography. Hepatol- 45. Cox ZL, Testani JM. Loop diuretic resistance complicating acute
ogy. 1994;19(2):312-321. heart failure. Heart Fail Rev. 2020;25(1):133-145.
25. Arroyo V, Cl aria J, Sal
o J, Jimenez W. Antidiuretic hormone and the 46. Cowie MR, Mosterd A, Wood DA, et al. The epidemiology of heart
pathogenesis of water retention in cirrhosis with ascites. Semin failure. Eur Heart J. 1997;18(2):208-225.
Liver Dis. 1994;14(1):44-58. 47. Griffin M, Soufer A, Goljo E, et al. Real World Use of Hyperton-
26. Perico N, Remuzzi G. Edema of the nephrotic syndrome: the role of ic Saline in Refractory Acute Decompensated Heart Failure: A
the atrial peptide system. Am J Kidney Dis. 1993;22(3):355-366. U.S. Center’s Experience. JACC Heart Fail. 2020;8(3):
27. Humphreys MH. Mechanisms and management of nephrotic 199-208.
edema. Kidney Int. 1994;45(1):266-281. 48. Wilcox CS, Mitch WE, Kelly RA, et al. Response of the kidney to
28. Siddall EC, Radhakrishnan J. The pathophysiology of edema for- furosemide. I. Effects of salt intake and renal compensation. J Lab
mation in the nephrotic syndrome. Kidney Int. 2012;82(6):635-642. Clin Med. 1983;102(3):450-458.

Adv in Kidney Disease and Health 2023;30(2):110-123


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en marzo 06, 2023. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
122 Koirala et al

49. Mullens W, Verbrugge FH, Nijst P, Tang WHW. Renal sodium avid- 69. Ter Maaten JM, Damman K, Hanberg JS, et al. Hypochloremia,
ity in heart failure: from pathophysiology to treatment strategies. Diuretic Resistance, and Outcome in Patients With Acute Heart
Eur Heart J. 2017;38(24):1872-1882. Failure. Circ Heart Fail. 2016;9(8):e003109.
50. Martens P, Dupont M, Verbrugge FH, et al. Urinary Sodium 70. Fonarow GC, Corday E, ADHERE Scientific Advisory Committee.
Profiling in Chronic Heart Failure to Detect Development of Acute Overview of acutely decompensated congestive heart failure
Decompensated Heart Failure. JACC Heart Fail. 2019;7(5):404-414. (ADHF): a report from the ADHERE registry. Heart Fail Rev.
51. Paterna S, Di Gaudio F, La Rocca V, et al. Hypertonic Saline in 2004;9(3):179-185.
Conjunction with High-Dose Furosemide Improves Dose-Response 71. Gamba G. WNK lies upstream of kinases involved in regulation of
Curves in Worsening Refractory Congestive Heart Failure. Adv ion transporters. Biochem J. 2005;391(Pt 1):e1-e3.
Ther. 2015;32(10):971-982. 72. Yang CL, Angell J, Mitchell R, Ellison DH. WNK kinases regulate
52. Licata G, Di Pasquale P, Parrinello G, et al. Effects of high-dose thiazide-sensitive Na-Cl cotransport. J Clin Invest. 2003;111(7):1039-
furosemide and small-volume hypertonic saline solution infusion 1045.
in comparison with a high dose of furosemide as bolus in refrac- 73. Piala AT, Moon TM, Akella R, He H, Cobb MH, Goldsmith EJ.
tory congestive heart failure: long-term effects. Am Heart J. Chloride sensing by WNK1 involves inhibition of autophosphory-
2003;145(3):459-466. lation. Sci Signal. 2014;7(324):ra41.
53. Paterna S, Fasullo S, Parrinello G, et al. Short-term effects of hyper- 74. Kahle KT, Ring AM, Lifton RP. Molecular physiology of the WNK
tonic saline solution in acute heart failure and long-term effects of a kinases. Annu Rev Physiol. 2008;70:329-355.
moderate sodium restriction in patients with compensated heart 75. Thorn CF, Ellison DH, Turner ST, Altman RB, Klein TE. PharmGKB
failure with New York Heart Association class III (Class C) summary: Diuretics pathway, pharmacodynamics. Pharmacogenet
(SMAC-HF Study). Am J Med Sci. 2011;342(1):27-37. Genomics. 2013;23(8):449-453.
54. Okuhara Y, Hirotani S, Naito Y, et al. Intravenous salt supplemen- 76. Moriguchi T, Urushiyama S, Hisamoto N, et al. WNK1 regulates
tation with low-dose furosemide for treatment of acute decompen- phosphorylation of cation-chloride-coupled cotransporters via the
sated heart failure. J Card Fail. 2014;20(5):295-301. STE20-related kinases, SPAK and OSR1. J Biol Chem.
55. Hanberg JS, Rao V, Ter Maaten JM, et al. Hypochloremia and 2005;280(52):42685-42693.
Diuretic Resistance in Heart Failure: Mechanistic Insights. Circ 77. Wilson FH, Kahle KT, Sabath E, et al. Molecular pathogenesis of in-
Heart Fail. 2016;9(8): e003180. https://doi.org/10.1161/CIRCHEART herited hypertension with hyperkalemia: the Na-Cl cotransporter
FAILURE.116.003180 is inhibited by wild-type but not mutant WNK4. Proc Natl Acad
56. Wesson DE. Glomerular filtration effects of acute volume expan- Sci U S A. 2003;100(2):680-684.
sion: importance of chloride. Kidney Int. 1987;32(2):238-245. 78. Casta~neda-Bueno M, Cervantes-Perez LG, Vazquez N, et al. Acti-
57. Urso C, Brucculeri S, Caimi G. Acid-base and electrolyte abnormal- vation of the renal Na1:Cl- cotransporter by angiotensin II is a
ities in heart failure: pathophysiology and implications. Heart Fail WNK4-dependent process. Proc Natl Acad Sci U S A.
Rev. 2015;20(4):493-503. 2012;109(20):7929-7934.
58. Briggs J. The macula densa sensing mechanism for tubuloglomeru- 79. Bazua-Valenti S, Chavez-Canales M, Rojas-Vega L, et al. The Effect
lar feedback. Fed Proc. 1981;40(1):99-103. of WNK4 on the Na1-Cl- Cotransporter Is Modulated by Intracel-
59. Kotchen TA, Luke RG, Ott CE, Galla JH, Whitescarver S. Effect of lular Chloride. J Am Soc Nephrol. 2015;26(8):1781-1786.
chloride on renin and blood pressure responses to sodium chloride. 80. Grodin JL, Sun JL, Anstrom KJ, et al. Implications of Serum Chlo-
Ann Intern Med. 1983;98(5 Pt 2):817-822. ride Homeostasis in Acute Heart Failure (from ROSE-AHF). Am J
60. Nashat FS, Tappin JW, Wilcox CS. Plasma sodium concentration Cardiol. 2017;119(1):78-83.
and sodium excretion in the anaesthetized dog. J Physiol. 81. Soleimani M. The multiple roles of pendrin in the kidney. Nephrol
1976;254(1):183-202. Dial Transplant. 2015;30(8):1257-1266.
61. Wilcox CS. Renal haemodynamics during hyperchloraemia in the 82. Patel-Chamberlin M, Varasteh Kia M, Xu J, Barone S, Zahedi K,
anaesthetized dog: effects of captopril. J Physiol. 1988;406:27-34. Soleimani M. The role of epithelial sodium channel ENaC and
62. Wilcox CS, Granges F, Kirk G, Gordon D, Giebisch G. Effects of sa- the apical Cl-/HCO3- exchanger pendrin in compensatory salt re-
line infusion on titratable acid generation and ammonia secretion. absorption in the setting of Na-Cl cotransporter (NCC) inactiva-
Am J Physiol. 1984;247(3 Pt 2):F506-F519. tion. PLoS One. 2016;11(3):e0150918.
63. Gordon D, Nashat FS, Wilcox CS. An analysis of the regulation of 83. Wall SM, Lazo-Fernandez Y. The role of pendrin in renal physi-
sodium excretion during induced changes in plasma sodium con- ology. Annu Rev Physiol. 2015;77:363-378.
centration in anaesthetized dogs. J Physiol. 1981;314:531-545. 84. Cil O, Haggie PM, Phuan PW, Tan JA, Verkman AS. Small-Mole-
64. Chowdhury AH, Cox EF, Francis ST, Lobo DN. A randomized, cule Inhibitors of Pendrin Potentiate the Diuretic Action of Furose-
controlled, double-blind crossover study on the effects of 2-L infu- mide. J Am Soc Nephrol. 2016;27(12):3706-3714.
sions of 0.9% saline and plasma-lyteÒ 148 on renal blood flow ve- 85. Wilcox CS, Sterzel RB, Dunckel PT, Mohrmann M, Perfetto M.
locity and renal cortical tissue perfusion in healthy volunteers. Ann Renal interstitial pressure and sodium excretion during hilar
Surg. 2012;256(1):18-24. lymphatic ligation. Am J Physiol. 1984;247(2 Pt 2):F344-F351.
65. Theisler CW. Chapter on Diuretics (Loop and Thiazide Diuretics), 86. Granger JP, Alexander BT, Llinas M. Mechanisms of pressure natri-
Adjuvant Medical Care. Boca Raton, FL: CRC Press; 2022. uresis. Curr Hypertens Rep. 2002;4(2):152-159.
66. Loon NR, Wilcox CS. Mild metabolic alkalosis impairs the natri- 87. Husain-Syed F, Gr€ one HJ, Assmus B, et al. Congestive nephropa-
uretic response to bumetanide in normal human subjects. Clin Sci thy: a neglected entity? Proposal for diagnostic criteria and future
(Lond). 1998;94(3):287-292. perspectives. ESC Heart Fail. 2021;8(1):183-203.
67. Ponce-Coria J, San-Cristobal P, Kahle KT, et al. Regulation of 88. Felker GM, Lee KL, Bull DA, et al. Diuretic strategies in patients
NKCC2 by a chloride-sensing mechanism involving the WNK3 with acute decompensated heart failure. N Engl J Med.
and SPAK kinases. Proc Natl Acad Sci U S A. 2008;105(24):8458- 2011;364(9):797-805.
8463. 89. Oppermann M, Hansen PB, Castrop H, Schnermann J. Vasodilata-
68. Peti-Peterdi J, Harris RC. Macula densa sensing and signaling tion of afferent arterioles and paradoxical increase of renal vascular
mechanisms of renin release. J Am Soc Nephrol. 2010;21(7):1093- resistance by furosemide in mice. Am J Physiol Renal Physiol.
1096. 2007;293(1):F279-F287.

Adv in Kidney Disease and Health 2023;30(2):110-123


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en marzo 06, 2023. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
Etiology and Management of Edema: A Review 123

90. Gilmer GG, Deshpande VG, Chou CL, Knepper M. Flow resistance 97. Hodson DZ, Griffin M, Mahoney D, et al. Natriuretic Response Is
along the rat renal tubule. Am J Physiol Renal Physiol. Highly Variable and Associated With 6-Month Survival: Insights
2018;315(5):F1398-F1405. From the ROSE-AHF Trial. JACC Heart Fail. 2019;7(5):383-391.
91. Simeoni M, Boyde A, Shirley DG, Capasso G, Unwin RJ. 98. Inoue M, Okajima K, Itoh K, et al. Mechanism of furosemide resis-
Application of red laser video-rate scanning confocal microscopy tance in analbuminemic rats and hypoalbuminemic patients. Kidney
to in vivo assessment of tubular function in the rat: selective action Int. 1987;32(2):198-203.
of diuretics on tubular diameter. Exp Physiol. 2004;89(2):181- 99. Chalasani N, Gorski JC, Horlander JC Sr, et al. Effects of albumin/
185. furosemide mixtures on responses to furosemide in hypoalbumine-
92. Fitch GK, Weiss ML. Activation of renal afferent pathways mic patients. J Am Soc Nephrol. 2001;12(5):1010-1016.
following furosemide treatment. II. Effect Of angiotensin blockade. 100. Fliser D, Zurbr€ uggen I, Mutschler E, et al. Coadministration of al-
Brain Res. 2000;861(2):377-389. bumin and furosemide in patients with the nephrotic syndrome.
93. Kopp UC. Role of renal sensory nerves in physiological and path- Kidney Int. 1999;55(2):629-634.
ophysiological conditions. Am J Physiol Regul Integr Comp Physiol. 101. Kitsios GD, Mascari P, Ettunsi R, Gray AW. Co-administration of
2015;308(2):R79-R95. furosemide with albumin for overcoming diuretic resistance in pa-
94. Araujo M, Solis G, Welch WJ, Wilcox CS. Renal Nerve Deafferen- tients with hypoalbuminemia: a meta-analysis. J Crit Care.
tation Attenuates the Fall in GFR during Intravenous Infusion of 2014;29(2):253-259.
Furosemide in Anesthetized Rats. Kidney Blood Press Res. 102. Martin GS, Mangialardi RJ, Wheeler AP, Dupont WD, Morris JA,
2020;45(1):70-83. Bernard GR. Albumin and furosemide therapy in hypoproteinemic
95. Almeshari K, Ahlstrom NG, Capraro FE, Wilcox CS. A volume-in- patients with acute lung injury. Crit Care Med. 2002;30(10):2175-
dependent component to postdiuretic sodium retention in humans. 2182.
J Am Soc Nephrol. 1993;3(12):1878-1883. 103. Haws RM, Baum M. Efficacy of albumin and diuretic therapy in
96. Cox ZL, Rao VS, Ivey-Miranda JB, et al. Compensatory post- children with nephrotic syndrome. Pediatrics. 1993;91(6):1142-1146.
diuretic renal sodium reabsorption is not a dominant mechanism 104. Kapur G, Valentini RP, Imam AA, Mattoo TK. Treatment of severe
of diuretic resistance in acute heart failure. Eur Heart J. edema in children with nephrotic syndrome with diuretics alone–a
2021;42(43):4468-4477. prospective study. Clin J Am Soc Nephrol. 2009;4(5):907-913.

Adv in Kidney Disease and Health 2023;30(2):110-123


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en marzo 06, 2023. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.

You might also like