You are on page 1of 13

193 Etiology and pathogenesis of gout

Tanya J. Major • Nicola Dalbeth

Key Points processes. However, pegloticase, which leads to dramatic reductions in serum
■ Gout is a chronic disease of monosodium urate crystal deposition, which typically urate concentrations, did not show increased lipid or protein oxidation, sug-
presents as recurrent episodes of severe, painful inflammatory arthritis. Monosodium gesting that urate is not a major factor controlling oxidative stress in vivo.2
urate crystals form from extracellular fluids saturated with urate, the endproduct of Urate may also have an important role in immune surveillance. Distressed
human purine metabolism. and dying cells produce locally high urate concentrations as a “danger” sig-
■ The gout flare is a severe but self-limited arthritis caused by an inflammatory response
nal, which acts as an endogenous adjuvant to help trigger inflammatory and
to monosodium urate crystals. Repeated gout flares and persistent crystal deposition
both innate and specific immune responses (discussed in more detail later).3
can lead to chronic gouty arthritis, tophi, and erosive gout.
Urate may also have had an evolutionary role in maintenance of blood pres-
sure and intravascular volume.4
■ Hyperuricemia (elevated serum urate concentration) is the central biochemical
precursor for gout. It is generally classified as a serum urate concentration exceeding
about 6.8 mg/dL (≈400 μM). FORMS OF URATE
■ Hyperuricemia results from an imbalance between urate production and excretion.
Urate exists in biologic systems in two forms with limited solubility, the ion-
In most patients with gout, hyperuricemia results from impairment of renal and gut
ized form (uric acid) and the unionized form (urate; Fig. 193.2). Uric acid
excretion.
(C5H4N4O3; 2, 6, 8-trioxypurine) is a weak organic acid ionized at position 9
■ Phagocytes from the synovial fluid of people with asymptomatic hyperuricemia and with a functional pKa1 in serum of 5.75. At the physiologic pH of most body
gout often contain monosodium urate crystals, without any overt inflammation, fluids (pH of 7.4), urate ion levels exceed those of unionized uric acid in a
indicating crystals in the synovial compartment do not always elicit an inflammatory ratio of about 50 to 1. In acidic body fluids (such as urine, pH of 5.0), union-
response. ized uric acid predominates. Because of the high concentration of sodium in
■ The gout flare is initiated by resident synovial cells, including phagocytes, which extracellular fluid, urate mainly functions as monosodium urate (MSU). As
secrete chemokines and cytokines to attract and activate the neutrophils that a consequence, the high solubility of the urate ion (≈120 mg/dL or 7.1 mM)
predominate in the synovial cavity of acutely inflamed joints. is replaced by the much lower solubility of monosodium urate (≈6.8 mg/dL
■ Activation of the NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) or 400 μM). Similarly, the solubility of uric acid is only about 10 to 15 mg/
inflammasome by monosodium urate crystals, leading to release of mature interleukin- dL (≈600–900 μM).
1β, is central to initiation of the gout flare. Serum urate concentrations exceeding 6.8 mg/dL are saturating, a con-
■ Termination of the gout flare is regulated by macrophage differentiation, release dition referred to as hyperuricemia. Persistent hyperuricemia reflects
of antiinflammatory soluble mediators, and formation of aggregated neutrophil extracellular fluid urate saturation. At these saturated levels, monosodium
extracellular traps (NETs) that degrade proinflammatory cytokines. Aggregated NETs urate monohydrate crystals are able to form and deposit in the joints and
may also play a role in tophus formation. other tissues. In the acidic environment of the urine, uric acid crystals and
■ Joint damage in advanced gout is strongly linked to intraarticular tophi, with stones (distinct from monosodium urate crystals) form in the urinary tract.
activation of catabolic pathways leading to focal cartilage and bone degradation. Increasing levels of hyperuricemia impart increasing risk for monosodium
■ Defects in purine metabolic enzymes (e.g., hypoxanthine-guanine urate crystal deposition and the associated clinical consequences.5
phosphoribosyltransferase and 5-phosphoribosyl 1-pyrophosphate synthetase 1) that Hyperuricemia is very common, affecting more than 20% of adult White
result in urate overproduction are well-characterized causes of gout but account for men in the United States,6 and is even more common among certain ethnic-
fewer than 1% of cases. ities. Although hyperuricemia most often does not evolve into clinical gout,
■ Large genetic studies have revealed many candidate genes associated with it is associated with several highly prevalent chronic disorders, as discussed
hyperuricemia and gout. These include genes involved in urate transport, metabolic in Chapter 192.
pathways, regulation of inflammation, and regulation of gene expression. Genetic
variations in the urate transporters SLC2A9 and ABCG2 are consistently and strongly
URATE PRODUCTION AND EXCRETION
associated with hyperuricemia and gout.
URATE BALANCE
Urate levels are maintained through a delicate balance between production
Gout is a chronic disease of monosodium urate (MSU) crystal deposition and excretion. This balance can be altered by many factors, including genet-
presenting as a painful and potentially destructive arthritis arising in the set- ics, environment, other clinical conditions, and medications (see Fig. 193.3
ting of hyperuricemia. Urate is the obligatory endproduct of human purine and Box 193.2). These factors alter both production and excretion, with
metabolism. The clinical manifestations of gout arise as a consequence of some having much larger influence than others.
monosodium urate crystal deposition and include the gout flare, chronic The bulk of urate usually derives from endogenous synthesis of purines
gouty arthritis, and tophi. This chapter focuses on the pathophysiology of from small-molecule nonpurine precursors, breakdown of dietary purines,
articular manifestations of gout, detailing the various “checkpoints” neces- and reutilization of preformed body purine compounds, as diet contains little
sary in developing clinically evident gout (Fig. 193.1). urate, and dietary urate is poorly absorbed.7 Urate production occurs largely
in the liver and to a minimal degree the small intestine. Urate released from
cells circulates relatively free (<4%) of serum protein binding7 so that all or
URATE PHYSIOLOGY nearly all circulating urate is filtered at the glomerulus. Under steady-state
The clinical features of gout occur as the result of inflammatory responses to conditions, urate production is balanced by excretion, largely through renal
monosodium urate crystals deposited because of one or more derangements excretion of uric acid (equivalent to about two thirds of daily production).
in the physiology of urate. In contrast to the case in most mammals, urate Urate secretion into the small intestine, with breakdown of urate by gut bac-
is the final product of purine metabolism in humans and higher primate teria (intestinal uricolysis), accounts for nearly all the rest of urate excretion.
species, in which the gene encoding the enzyme uricase (urate oxidase) has The body pool of urate is expanded in hyperuricemic states resulting from
been silenced by loss-of-function genetic variants.1 impaired urate excretion or urate overproduction.
Urate is the most abundant natural antioxidant in the human body Urate pools in normal men range from about 800 to 1500 mg and in
(Box 193.1). The traditional view has been that a major physiologic role of women from 500 to 1000 mg. Daily turnover of the body’s urate pool (bal-
urate is to remove reactive oxygen species (ROS), possibly providing pro- anced production and excretion) is considerable, averaging 0.6 to 0.7 pools
tection against oxidant-induced neurologic and cardiovascular degenerative (≈0.5 to 1 g) every day.

1700
F 2 A 2 2, A 2 2 2 A D D 2C ,A2 2AA @DI 1 E A C A ) 2. 3 E A
, 3AD2A , A A 2 D 0 C AD F C DC A ) A C E A ( A C A AE
CHAPTER 193 Etiology and pathogenesis of gout 1701

GOUT CHECKPOINTS, DISEASE STAGES, AND INFLUENCING FACTORS

“Check points” Disease Stages Influencing Factors

Normouricemia
Genetics, environment,
Increased serum urate medications, clinical features
(overproduction and/or (see Box 193.2)
underexcretion)

Hyperuricemia

Seed nucleus, temperature,


Crystal formation
pH (see Box 193.3)

Monoscdium urate
crystal deposition

Acute inflammatory
Genetics, crystal coating/size,
response initiated by NLRP3
trigger events (see Box 193.3)
inflammasome activation

Recurrent gout flares

Chronic inflammatory Genetics, kidney function,


response/tophus formation sustained hyperuricemia

Chronic gouty arthritis,


tophaceous gout, gouty
bone erosion

FIG. 193.1 There are a number of “checkpoints” involved in the development and progression of gout: (1) development of hyperuricemia; (2) formation and deposition of
monosodium urate crystals; and (3) clinical manifestations of gout (gout flares, chronic gouty arthritis, and tophaceous gout).

BOX 193.1 PHYSIOLOGIC FUNCTIONS OF PURINES


FACTORS AFFECTING URATE BALANCE
Antioxidation (urate)
Nucleotide building blocks for nucleic acids (DNA and RNA)
Nucleotide components of coenzymes (e.g., coenzyme A, FAD, NAD) Renal tubular reabsorption (e.g., URAT1, GLUT9)
High-energy phosphate donors in enzyme reactions (e.g., ATP) Dietary purines, alcohol
Metabolic regulators (e.g., cyclic AMP, cyclic GMP) Metabolic disorders, insulin resistance
Neurotransmitters Purine salvage pathways
Extracellular messengers (e.g., adenosine, ATP) ATP turnover (e.g., ethanol, fructose, illness)
Intestinal excretion
Intracellular second messengers (e.g., G protein–coupled receptors) (mediated by ABCG2)
Raise
Immunologic adjuvants, endogenous “danger” signal for innate immunity Glomerular filtration
urate pool
Maintenance of extracellular volume, vascular tone (urate) (hypothesized) Urate-lowering drugs, diet
Weight reduction
AMP, Adenosine monophosphate; ATP, adenosine triphosphate; FAD, flavin adenine dinucleotide; GMP, Renal tubular secretion
guanosine monophosphate; NAD, nicotinamide adenine dinucleotide. (e.g., ABCG2, NPT1, MRP4)

Lower
urate pool
URIC ACID AND THE URATE ANION

O O
H H
C6 N C6 N
HN1 5 C
7
pKa1 = 5.75 HN1 5 C
7
FIG. 193.3 The systemic urate pool and the likelihood of gout are determined by
+
2 8 C O 2 8 C O+H the dynamic balance between endogenous synthesis and recycling of purines and
O C 3
4
C 9 O C 3
4
C 9
N N N N– excretion by the kidney and gut. ABCG2, ATP-binding cassette subfamily G member
H H H 2; ATP, adenosine triphosphate; GLUT9, glucose transporter 9; MRP4, multidrug
resistance protein 4; NPT1, sodium phosphate transport protein 1; URAT1, urate
Uric acid Urate anion
transporter 1.

FIG. 193.2 At physiologic pH (7.4), urate predominates at about 50 : 1 over union-


ized uric acid. Urate ion solubility is functionally reduced at the high sodium concen- results in less renal tubular uric acid reabsorption and thus increased uric
tration of extracellular fluids, so monosodium urate and uric acid have relatively low acid clearance in women. With the onset of menopause, serum urate values
solubilities in biologic fluids. in women increase and approach or equal those of men.
Urate saturation of extracellular fluids (for which hyperuricemia is a
surrogate marker) predisposes to monosodium urate crystal formation and
Serum urate concentrations in children are lower than those in adults. deposition, events required for clinical expression of gout. The natural dif-
During male puberty, values increase into the adult male range. Serum urate ferences in serum urate levels between men and women explain the greater
levels remain lower in women of reproductive age than in their male coun- prevalence of gout in men. Additionally, the increase in serum urate levels
terparts.8 This gender difference results mostly from the action of estrogens,9 in postmenopausal women is accompanied by increases in the incidence of
which alter the activity of key urate transporter proteins in the kidney. This hyperuricemia and, over the succeeding 2 to 3 decades, gout.

F 2 A 2 2, A 2 2 2 A D D 2C ,A2 2AA @DI 1 E A C A ) 2. 3 E A


, 3AD2A , A A 2 D 0 C AD F C DC A ) A C E A ( A C A AE
1702 SECTION 17 Crystal-Related Arthropathies

BOX 193.2 FACTORS ASSOCIATED WITH URATE VARIATION


Genetic Factors Environmental Influences Clinical Associations
Male sex Associated with increased urate Older age
Common variants associated with hyperuricemia ■ Fasting/starvation Postmenopause in women
(replicated) ■ High purine foods (e.g., red meat, liver, offal, Medical comorbidities
■ SLC2A9 shellfish) ■ Hemolytic disorders
■ ABCG2 ■ Dehydration ■ Hemopoietic malignancies; tumor lysis
■ PDZK1 ■ Lead exposure ■ Lactic or ketoacidosis; hypoxemic states
■ GCKR ■ Fructose- and sugar-sweetened drinks ■ Lead nephropathy, chronic low-level exposure
■ RREB1 ■ Alcohol (beer and spirits) ■ Preeclampsia
■ SLC17A3 Associated with lower urate ■ Renal impairment
■ SLC16A9 ■ Low-fat dairy products ■ Psoriasis
■ SLC22A11 ■ DASH diet ■ Vasopressin-resistant diabetes insipidus
■ SLC22A12 ■ Mediterranean diet ■ Bartter and Gitelman syndromes
■ INHBC ■ Cherries ■ Down syndrome
Rare monogenic causes ■ Vitamin C ■ Hypertriglyceridemia
■ HPRT deficiencies: complete (Lesch-Nyhan ■ Coffee ■ Hypertension
syndrome), partial (Kelley-Seegmiller syndrome) ■ Obesity
■ PRPP synthetase overactivity ■ Cardiovascular disease
■ Glucose-6-phosphatase deficiency Medications associated with hyperuricemia
■ Fructose-1-P aldolase deficiency ■ Aspirin (low dose)
■ Myogenic glycogenoses (types III, V, VII) ■ Chemotherapeutic cytotoxics
■ Uromodulin-associated kidney disease: FJHN, ■ Diuretics
MCKD types 1 and 2 ■ Pyrazinamide
■ Ethanol
■ Levodopa
■ Nicotinic acid
■ Cyclosporine and tacrolimus
Medications associated with reduced serum urate
■ Losartan
■ Fenofibrate
■ Leflunomide
■ Calcium channel blockers
■ Atorvastatin
■ Sevelamer

FJHN, Familial juvenile hyperuricemic nephropathy; HPRT, hypoxanthine-guanine phosphoribosyltransferase; MCKD, medullary cystic kidney disease; PRPP, 5-phosphoribosyl 1-pyrophosphate.

PURINE METABOLISM AND URATE PRODUCTION oxidase form (xanthine oxidase [XO]) that uses oxygen (O2) to convert
hypoxanthine and xanthine to urate and a dehydrogenase form (XDH) that
Urate is the endproduct of purine metabolism in humans and other great uses the nucleotide NAD+. Inhibition of XOR activity is the major action of
apes, as well as birds and reptiles. Purines are compounds possessing the the most common class of agents used for urate lowering in patients with
nine-member purine nucleus composed of fused pyrimidine and imidazole gout.11 Other roles for xanthine oxidoreductase have been proposed, includ-
rings. Purines fulfill essential functions in all living cells (see Box 193.1). ing protection from infection and tissue injury (caused by the generation of
Most functions of purines are carried out by nucleotide and nucleo- unstable oxygen radicals) during and after ischemia.
side derivatives of the purine bases adenine, hypoxanthine, and guanine. Purine nucleotide synthesis and degradation are both carefully regulated
Representative structures of purine bases, nucleosides, and nucleotides are processes (Fig. 193.6).10 Malfunctions in this regulatory process lead to
shown in Fig. 193.4. increased cellular levels of PRPP, resulting in accelerated purine nucleotide
The biochemical pathways of purine metabolism and their regulation are and urate production. This occurs in two rare X chromosome–linked disor-
discussed in more detail elsewhere.10 The only endogenous pathway of net ders (discussed later).10 Similarly, excessive cellular adenosine triphosphate
purine production is called purine synthesis de novo and involves 10 steps (ATP) depletion (e.g., in tissue hypoxia or acute alcohol intoxication) can
(Fig. 193.5). The purine ring is synthesized on a backbone of ribose-5-phos- lead to reduced inhibitory nucleotide concentrations and consequent urate
phate donated by the key regulatory substrate, 5-phosphoribosyl 1-pyrophos- overproduction. Other states of excessive urate production among patients
phate (PRPP) from the pentose phosphate sugar pathway. The endproduct with gout are not as well characterized at the pathway level.
of this pathway is the nucleotide inosine monophosphate (IMP). IMP serves
as a branch point in the conversion of new purines into the adenylate and
guanylate classes of purine nucleotides or, alternatively, into the purine deg-
URATE EXCRETION
radation pathway, culminating in urate formation. Urate excretion is mediated by multiple transporter proteins in the renal
Purine synthesis de novo is an energy costly process. Considerable sav- proximal tubule and the intestinal mucosa. The majority of urate is disposed
ing in cellular energy expenditure is achieved by an extensive network of of through the renal pathway (~60%), with the remaining ~30% being dis-
reactions that interconvert and salvage purine nucleotides, nucleosides, and posed of via the intestinal pathway. The sum of urate excretion via these
bases. This saves energy and provides flexibility in the provision of specific two pathways almost makes up the total urate clearance, and each pathway
purines to a wide array of cellular functions. can compensate, to a small extent, for underexcretion by the other pathway.
Degradation of purine nucleotides and nucleosides to purine bases cul-
minates in the key urate precursors, hypoxanthine and guanine. These Extrarenal urate excretion
are mostly reused in salvage reactions with PRPP, catalyzed by the enzyme Urate excretion in the intestines is primarily mediated by ABCG2, an ATP-
hypoxanthine-guanine phosphoribosyltransferase (HPRT). The remainder binding cassette (ABC) transporter, encoded by the gene ABCG2. ABCG2
of guanine is deaminated to xanthine. Unsalvaged hypoxanthine is oxidized functions in both the kidney and intestine, and dysfunction is associated
to xanthine, which undergoes further oxidation to urate. The enzyme xan- with hyperuricemia and gout.12 Analysis of the role of ABCG2 in intestinal
thine oxidoreductase (XOR) catalyzes both the conversions of hypoxanthine handling of urate has challenged the conventional concept of hyperurice-
to xanthine and xanthine to urate. mia caused by urate overproduction.13 Abcg2 knockout mice have reduced
Xanthine oxidoreductase is a molybdopterin cofactor and iron sulfide intestinal urate excretion, leading to higher serum urate levels. Despite also
cluster-containing flavoprotein that exists in interconvertible forms: an lacking Abcg2 in the kidneys, renal uric acid excretion in this situation is

F 2 A 2 2, A 2 2 2 A D D 2C ,A2 2AA @DI 1 E A C A ) 2. 3 E A


, 3AD2A , A A 2 D 0 C AD F C DC A ) A C E A ( A C A AE
CHAPTER 193 Etiology and pathogenesis of gout 1703

PURINE STRUCTURES

Purine base (e.g., adenine) Urate anion Pyrimidine base (e.g., thymine)

NH2 OH H

C6 N7 C N C4
N1 C5 N C H N3 C5 CH3

C8 H C O
H C2 C4 HO C C O C2 C6 H

N3 N9 N N N1

H H

Nucleoside (e.g., adenosine) Nucleotide (e.g., dATP)

NH2 NH2

2–
HOCH2 PO4 PO3– PO3– OCH2
5’ 4’ 1’
3’ 2’
HO OH HO H

FIG. 193.4 Purine ring atoms are numbered 1 to 9 as shown in the upper left panel. Pentose sugar carbon atoms are numbered 1′ to 5′, as shown in the lower left panel. In
purine nucleosides, a purine base is joined to a pentose ring through an N-glycoside bond between the purine 9 and pentose 1′ atoms. Nucleotides are phosphate esters of
the nucleoside, containing one, two, or three phosphate groups (nucleoside mono-, di-, or triphosphates, respectively) attached at the 5′ carbon of the sugar. Nucleotidases
remove phosphate groups. Phosphorylases remove both the phosphate group(s) and the sugar. Kinases transfer a high-energy phosphate group (usually donated by ade-
nosine triphosphate). The nucleoside of hypoxanthine is known as inosine, and the respective nucleotide is inosine monophosphate. dATP, Deoxyadenosine triphosphate.

PURINE SYNTHESIS, INTERCONVERSION, AND DEGRADATION IN HUMANS

Ribose-5-P, Mg-ATP

PRS
DNA, RNA DNA, RNA
PRPP

ATP GTP
10 steps

AMP AMPD IMP GMP

ND ND ND

APRT Adenosine ADA Inosine HPRT Guanosine HPRT

PNP PNP

Adenine Hypoxanthine Guanine

XO

Xanthine

XO

Urate

FIG. 193.5 The gene for uricase (urate oxidase) has been inactivated in humans and other primate species, so that urate is the end-product of purine metabolism. Intermediates
and enzymes not pertinent to hyperuricemia and gout have been omitted from this figure for simplicity. ADA, Adenosine deaminase; AMPD, AMP deaminase; APRT, adenine
phosphoribosyltransferase; GMP, guanosine monophosphate; ND, 5′-nucleotidase; PNP, purine nucleoside phosphorylase; XMP, xanthosine monophosphate; XO, xanthine
oxidase (oxidoreductase). For other abbreviations, see text.

F 2 A 2 2, A 2 2 2 A D D 2C ,A2 2AA @DI 1 E A C A ) 2. 3 E A


, 3AD2A , A A 2 D 0 C AD F C DC A ) A C E A ( A C A AE
1704 SECTION 17 Crystal-Related Arthropathies
still higher than normal, indicating that other transporters in the kidney can
REGULATION OF PURINE NUCLEOTIDE SYNTHESIS partially compensate.
Reduced extrarenal urate excretion leads to an increase in the renal excre-
ATP + Rib-5-P tion load, which gives the appearance of urate overproduction. Thus it has
been suggested that urate overproduction may be better described as “renal
– PRPP overload,” consisting in turn of “extrarenal underexcretion” and “genuine
synthetase urate overproduction” subtypes.13
PRPP
Renal urate excretion
+ Despite nearly complete filtration of urate at the glomerulus, renal uric acid
– clearance in adults averages only 5% to 10% that of creatinine clearance,
Amido-PRT reflecting net renal tubular reabsorption of about 90% of filtered uric acid.
Adenine Hypoxanthine Within the renal tubule, uric acid reabsorption and secretion are regulated
APRT HPRT Guanine by a series of transporter proteins that ultimately regulate serum urate
concentrations.
Most people with hyperuricemia resulting from impaired renal uric acid
excretion show normal amounts of uric acid in the urine but have selectively
reduced uric acid clearance.14,15 Excretion of “normal” amounts of uric acid
Purine is accomplished by patients with gout only when serum urate levels are 2 to
nucleotides 3 mg/dL (120 to 180 μM) higher than in healthy people excreting the same
amounts of uric acid. The hyperuricemia that results from this is the prime
risk factor for developing gout.
FIG. 193.6 The major purine synthetic steps targeted by regulation are (1) the syn- Many mechanisms contribute to net uric acid excretion. Molecular and
thesis of PRPP in the PRPP synthetase (PRS) reaction and (2) the utilization of genomic approaches have identified several transporters involved in renal
PRPP in the first step of purine synthesis de novo. Rates of the 10-step de novo uric acid excretion, including some with substantial specificity for uric
pathway of purine nucleotide synthesis (vertical arrows) are regulated by the activ- acid.16 Alterations in uric acid movement may be caused by changes in these
ity of the enzyme AmidoPRT, which catalyzes the first step. AmidoPRT activity is urate transporters, changes in associated proteins or ion cotransporters, or
allosterically controlled by the antagonistic interaction of pathway purine nucleotide regulation of transport function. Two of these urate transporters, glucose
products that inhibit (−) the enzyme and the regulatory substrate 5-phosphoribosyl transporter 9 (GLUT9) and urate transporter 1 (URAT1), have strong effects
1-pyrophosphate (PRPP) that activates (+) AmidoPRT. Purine nucleotides also on serum urate levels (Fig. 193.7).
inhibit (−) PRPP synthetase, the enzyme-catalyzing synthesis of PRPP. Regulation
directed at these sequential reactions provides a flexible means to control purine Urate transporters
nucleotide production in an economical manner, a process potentiated by preferen- Glucose transporter 9
tial single-step salvage of preformed purine bases in reactions with PRPP catalyzed Glucose transporter 9 (GLUT9) is the product of the SLC2A9 gene. It is a
by the phosphoribosyltransferase enzymes hypoxanthine-guanine phosphoribosyl- voltage-driven urate transporter that mediates uric acid reabsorption from
transferase (HPRT) and APRT (depicted by the curved arrows). APT, Adenosine the tubular cell to the circulation. Glucose transporter 9 also transports the
triphosphate; Rib-5-P, ribose-5-phosphate. sugars glucose and fructose. In humans, GLUT9 exists in two isoforms:

RENAL HANDLING OF URATE

Proximal tubule lumen

Apical brush border

NPT1 MRP4 ABCG2 URAT1 GLUT9 Others Proximal


tubular
cell

Basolateral membrane

OATs and others GLUT9

Peritubular capillary

FIG. 193.7 The key players in reabsorption and secretion of uric acid across the proximal tubule epithelial cell. Exchange of uric acid for other anions is mediated by specialized
channels and transport proteins embedded in the tubular cell membrane. Whereas urate transporter 1 (URAT1) and glucose transporter 9 (GLUT9) are significant contributors
to uric acid absorption (such that defective function is associated with hypouricemia), ATP-binding cassette subfamily G member 2 (ABCG2), sodium-dependent phosphate
transporter protein 1 (NPT1), and multidrug resistance protein 4 (MRP4) are associated with net uric acid excretion. Uric acid transport is driven in part by a pH gradient pro-
duced by active sodium–hydrogen ion exchange. Several other organic anion transporters (OATs) have been implicated in uric acid transport. Transporters may be linked with
each other and with regulatory and signaling elements by PDZK1 scaffolding proteins, comprising the urate transportasome. Many of the drugs and endogenous mediators that
affect renal uric acid disposition interact with these proteins (e.g., the uricosuric drugs probenecid, benzbromarone, and lesinurad inhibit uric acid reabsorption by URAT1).

F 2 A 2 2, A 2 2 2 A D D 2C ,A2 2AA @DI 1 E A C A ) 2. 3 E A


, 3AD2A , A A 2 D 0 C AD F C DC A ) A C E A ( A C A AE
CHAPTER 193 Etiology and pathogenesis of gout 1705

GLUT9L, identified on the basolateral aspects (“blood” side) of the prox- unable to fully compensate for each other, resulting in lower total uric acid
imal renal tubular epithelial cell, and GLUT9S on the apical (“urine”) excretion and higher serum urate levels overall.
side.17–19
Glucose transporter 9 is also expressed in the basolateral membrane of
hepatocytes and regulates serum urate concentrations through dual roles in
CLASSIFICATION OF HYPERURICEMIA
uric acid handling in the kidney and uptake in the liver.20 Mice with systemic Hyperuricemia and gout may arise in the presence or absence of an under-
knockout of Slc2a9 (Glut9) have moderate hyperuricemia, massive hyper- lying clinical disorder or toxin or drug exposure. When a specific process
uricosuria, and an early-onset nephropathy. In contrast, specific inactivation resulting in hyperuricemia is identifiable, it is said to be secondary. When
of the Slc2a9 gene in the livers of adult mice leads to severe hyperuricemia no such process is evident, hyperuricemia is termed primary or idiopathic.
and hyperuricosuria, without renal disease.20 Several diseases, toxic states, and medications lead to hyperuricemia as a
result of urate overproduction (see Box 193.2). Foremost among these are
Urate transporter 1 diseases associated with cellular proliferation and destruction, such as acute
Urate transporter 1 (URAT1) is encoded by the SLC22A12 gene and has a leukemias and lymphomas, tumor lysis syndromes, hemolytic states, and
typical organic anion transporter structure.21 URAT1 is highly specific for psoriasis.
uric acid. URAT1 mediates the exchange of uric acid for a variety of endog- The causes of hyperuricemia have traditionally also been divided into
enous and drug anions known to affect renal uric acid transport. Similar to overproduction causes (mainly comprising metabolic factors) and underex-
GLUT9S, URAT1 localizes to the apical brush border membrane of proximal cretion causes (mostly renal). Many patients with gout have a combination
tubular epithelial cells and is involved in the reabsorption of uric acid from of underexcretion and overproduction causes, with relative uric acid under-
the lumen. The uricosuric drugs probenecid, benzbromarone, and lesinurad excretion by the kidneys contributing the most to hyperuricemia.
increase uric acid excretion by inhibiting URAT1 and other OATs.21,22
URAT1 interacts with the scaffolding protein PDZK1. PDZ motifs are
typically involved in protein–protein interactions that support intracellular
MONOSODIUM URATE CRYSTAL FORMATION
signaling and are also present in other urate transporters, including OAT4 The formation of monosodium urate crystals requires sustained high (super-
and sodium phosphate transport protein 1 (NPT1, discussed below). This saturated) concentrations of urate. Crystal formation is influenced by many
suggests that a more extensive multiprotein complex, the “urate transpor- other factors, including the presence of particulate seed nuclei such as carti-
tasome” (containing transporters, transport regulatory molecules, hormone lage debris, collagen, chondroitin, and hyaluronate released by joint trauma
receptors, and intracellular signaling elements), may be involved in regu- or osteoarthritis (OA); local cation concentrations; pH, temperature, and
lating bidirectional uric acid transport across the renal tubular epithelial dehydration; and the balance between macromolecular inhibitors and pro-
cell. The fractional renal excretion of Uric acid (FEua = uric acid clearance/ moters of crystal formation (Box 193.3).28 Immunoglobulin G (IgG) and
creatinine clearance × 100%) in Slc22a12 (Urat1) knockout mice23 remains immunoglobulin M (IgM) may also promote crystal formation in patients
substantially less than 100%. This confirms that there are multiple mecha- with gout by providing a stable molecular platform for crystal nucleation
nisms of uric acid reabsorption. and growth.29,30 Monosodium urate crystals preferentially form at certain
sites, particularly the first metatarsophalangeal joint, midfoot, and Achilles
Other urate transporters tendon.31
Many other urate transporters are known. The sodium-dependent phosphate There is growing evidence to suggest a link between the presence of osteo-
cotransporters type 1 and type 4 (NPT1 and NPT4; encoded by SLC17A1 and arthritis and sites of monosodium urate crystal deposition.32 A cadaveric
SLC17A3, respectively) are located at the apical membrane of the proximal study reported that monosodium urate crystals were nearly always located
tubule and mediate net tubular uric acid secretion.24 Multidrug resistance within or adjacent to an osteoarthritic cartilage lesion.33 The osteoarthritic
protein 4 (MRP4; encoded by ABCC4) is another renal apical organic anion
efflux transporter that may contribute to tubular secretion of uric acid.25 In
addition, ABCG2 is also expressed on the apical surface of the renal tubule
BOX 193.3 GOUT PROMOTERS AND INHIBITORS
and might play a role in secretion of uric acid into the tubular lumen, as well
as its role within the gut.26 Crystal formation
The relative importance of the many other OATs and exchangers in Seed nucleus (particulate)
human renal uric acid handling is also uncertain. For example, OAT4, Immunoglobulin
encoded by SLC22A11 and localized at the apical membrane of the proximal Phagocytes
tubules, is also associated with serum urate level,27 and OAT10 is a second- Low temperature
ary target for some uricosuric drugs. Low pH
Cation concentration
Intraarticular dehydration
HYPERURICEMIA Other (unknown) macromolecules
CONDITIONING FACTORS Triggering the gout flare (local factors)
Several genetic and physiologic circumstances predispose all humans to Rapid change in urate level
the development of hyperuricemia and gout, namely: the species-wide defi- Crystal release
ciency of uricase, the enzyme catalyzing conversion of urate to the much IgG coat (apolipoproteins B, E inhibitory)
more soluble excretory product allantoin; net reabsorption of 90% of urate Complement activation (classical, alternate, MAC)
filtered at the glomerulus; and the limited solubility of both urate and mono- NLRP3 inflammasome activation
sodium urate in body fluids. Cytokine and chemokine release
Despite these conditioning factors, most people do not display the sus- Endothelial activation (e-selectin, ICAM-1, VCAM-1)
tained hyperuricemia that reflects extracellular fluid saturation, and even Local trauma (?)
fewer develop monosodium urate crystal deposition or gout. Studies into
the influences of serum urate levels have identified a number of factors asso- Presence of susceptible phagocytes, mast cells (systemic events)
ciated with increased or decreased urate, including genes, environmental Surgery, trauma
or clinical factors, and medications (Fig. 193.3; see also Box 193.2). For Infections, other intercurrent systemic illness
some of these associations, the causal relationship and underlying mecha- Alcohol, dietary intake
nisms have been determined (e.g., lead poisoning–induced kidney failure), Drugs that raise or lower circulating urate level
whilst others have less certain mechanisms (e.g., reduction in urate levels
associated with low-fat dairy consumption). The many processes influenc- A diverse array of proteins and other mediators have been identified on the surfaces of monosodium
urate crystals. In addition to their proinflammatory effect through opsonizing existing crystals,
ing development of hyperuricemia operate through impaired renal uric acid
immunoglobulin M (IgM) and immunoglobulin G (IgG) antibodies may promote crystal formation by
excretion (most commonly), excessive urate production, reduced extrarenal providing a stable molecular platform for crystal nucleation and growth. Apolipoproteins are the best
excretion, or a combination of these mechanisms.14 characterized of the antiinflammatory molecules that coat crystals. The characteristics of the phagocytes
With increased urate production or decreased renal uric acid excretion, encountering the crystals may be crucial; macrophages that are more differentiated are less likely to
elicit proinflammatory cytokines.
intestinal uricolysis increases. On the other hand, when intestinal urate
ICAM-1, Intercellular adhesion molecule 1; MAC, membrane attack complex; VCAM-1, vascular cell
excretion is impaired, the kidneys are confronted with a higher urate load.7 adhesion molecule 1.
In individuals with hyperuricemia these two excretory mechanisms are

F 2 A 2 2, A 2 2 2 A D D 2C ,A2 2AA @DI 1 E A C A ) 2. 3 E A


, 3AD2A , A A 2 D 0 C AD F C DC A ) A C E A ( A C A AE
1706 SECTION 17 Crystal-Related Arthropathies
joint has increased levels of cartilage degradation products present, such as Although the inflammation that monosodium urate crystals trigger via
chondroitin sulphate, which lowers urate solubility and promotes nucle- the inflammasome is clearly detrimental in the context of the gout flare,
ation and growth of monosodium urate crystals.34–36 In addition, the pres- it may play a physiologic role in activating the innate immune response
ence of type 2 cartilage from osteoarthritic joints encourages the formation by providing a “danger” signal.3 Danger signals from monosodium urate
of smaller monosodium urate crystals. These smaller crystals have a higher crystals and from other purines (e.g., ATP) may also help trigger the
inflammatory potential than larger crystals.37 adaptive immune response by activating antigen-presenting dendritic
One reason why most people with persistent hyperuricemia do not get cells.3 The dendritic cells provide stimulatory signals that help drive
gout may lie in individual variation in the propensity for forming crystals. the activation of T lymphocytes, including type 17 helper T cell (Th17)
However, microscopic and imaging studies have shown that many people differentiation.
with asymptomatic hyperuricemia have evidence of monosodium urate crys-
tals within joints,38 suggesting that crystals may be present within the joint
for long periods of time without eliciting an overt inflammatory response
NLRP3 INFLAMMASOME ACTIVATION
leading to the presentation of gout. The NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflam-
masome is a multiprotein cytoplasmic complex that senses cell damage and
the presence of microbial products.44 It consists of a sensor protein: NACHT,
PROTEINS BINDING TO MONOSODIUM URATE CRYSTALS LRR, and PYD domains containing protein 3 (NALP3, also known as cryopy-
Monosodium urate crystals are composed of a highly ordered, regular array rin, encoded by the NLRP3 gene), an adaptor protein: apoptosis-associated
of urate and sodium ions and water molecules. Although the monosodium speck-like protein containing a CARD (ASC, also known as PYCARD), and
urate crystal surface has a net negative charge, some positive charges are an effector enzyme: caspase 1.44 Assembly of the NLRP3 inflammasome leads
also exposed. Monosodium urate crystal surfaces can bind a variety of lipids, to the release of two proinflammatory cytokines, IL-1β and IL-18, resulting
lipoproteins, and proteins. These proteins and lipids binding the surface of in an acute inflammatory cascade. Activation of the NLRP3 inflammasome
monosodium urate crystals may change the inflammatory potential of crys- is central to the gout flare, and clinical trials have reported efficacy of IL-1
tals. For example, monosodium urate crystal binding of immunoglobulin G antagonists such as anakinra and canakinumab in both treating and prevent-
(IgG), which attaches through both charge interactions and hydrogen bond- ing gout flares.
ing, results in a conformational change in IgG, encouraging inflammation The activation of the NLRP3 inflammasome involves two signaling path-
via phagocytosis by cells with Fcγ receptors (e.g., FcγRIII, CD16). ways, a priming signal via toll-like receptors and an assembly signal from
Apolipoprotein coating of monosodium urate crystals can counter the monosodium urate crystals. This two-signal system may explain the spo-
effects of IgG by decreasing the attractiveness of monosodium urate crystals radic nature of gout flares.
to phagocytes.39 ApoB is a major component of the serum low-density lipo-
protein fraction and can coat crystals to inhibit stimulation of neutrophils. Priming signal
ApoE has similar effects and can be made locally by synovial macrophages. The NLRP3 inflammasome priming signal controls the production of the
Other proteins detected on monosodium urate crystal surfaces include sensor, adaptor, and effector proteins that make up the inflammasome com-
further complement activation products, lysosomal enzymes, and the anti- plex. This signal is mediated by toll-like receptor (TLR) proteins, in partic-
inflammatory cytokine transforming growth factor-β (TGF-β). The inflam- ular TLR-2 and TLR-4. These TLR proteins do not seem to directly interact
matory potential of monosodium urate crystals reflects the balance between with monosodium urate crystals, but with other inflammatory mediators or
pro- and antiinflammatory elements of this coating. external factors.45
During phagocyte cell activation the S100 protein heterodimer S100A8/9,
formed by the calgranulin proteins S100A8 and S100A9 (formerly myeloid
THE GOUT FLARE release proteins MRP8 and MRP14), is secreted. The S100A8 and S100A9
A gout flare occurs when monosodium urate crystals interact with host cells proteins are endogenous ligands of TLR-4 and are among the earliest inflam-
to initiate an acute inflammatory response.40 Despite potent proinflamma- matory mediators produced in the response to monosodium urate crystals.
tory potential, monosodium urate crystals do not necessarily trigger acute The S100A8/9 heterodimer is also elevated in synovial fluid and serum
inflammation. Patients with gout and healthy hyperuricemic individuals during the gout flare, indicating it may be an important initial stimulus to
may have monosodium urate crystals in clinically uninvolved joints. Heavily attract and activate other inflammatory cells.46
crystal-laden fluids (“urate milk”) are sometimes found in relatively unin- The toll-like receptor TLR-2 may mediate the priming signal in gouty
flamed bursae and joints. The dense masses of monosodium urate crystals inflammation through its interaction with long-chain free fatty acids.47
in tophi (described in detail later) can reach massive dimensions often with Research into NLRP3 inflammasome activation by monosodium urate crys-
little apparent inflammatory response. In confined spaces, such as in axial tals found crystals alone were unable to stimulate an inflammatory response,
or visceral sites, tophi can enlarge progressively without eliciting symptoms but when combined with long-chain free fatty acids an inflammatory
until there is critical compression of neighboring tissues. response was observed.48 This response was mediated by TLR-2.
Similarly, patients with intercritical and advanced gout may have pro- Once the toll-like receptor proteins have interacted with an inflam-
longed periods with absent or relatively low-grade synovitis despite the matory mediator or other factor the priming signal pathway proceeds via
ongoing presence of monosodium urate crystals in the synovial environ- myeloid differentiation factor 88 (Myd88) and a chain of intracellular
ment. Furthermore, synovial fluid phagocytes from noninflamed joints of mediators (see Box 193.4)48 that converge on nuclear factor-κB (NF-κB),
patients with gout often contain monosodium urate crystals.41 This indicates allowing the production of the NLRP3 inflammasome components. Other
that crystals in the synovial compartment do not necessarily elicit overt adaptor molecules in the TLR-2 and TLR-4 signaling pathways (e.g., CD14)
inflammation despite active engagement with phagocytes. Thus whether may also play a role.
monosodium urate crystals initiate clinically significant inflammation
depends on multiple factors, including crystal size, the proteins and other Assembly signal
molecules coating them, the local cytokine milieu, and which cells they first Monosodium urate crystals have a strong surface reactivity, enabling the
encounter. crystals to cross-link membrane glycoproteins and directly activate proin-
In the gout flare, the predominant inflammatory cell in the synovial flammatory signaling pathways at several levels within the phagocyte.49 The
fluid and synovial membrane is the neutrophil, and activation/recruit- crystals interact directly with signaling proteins in the cell membrane (see
ment of neutrophils seems to contribute the bulk of the proinflammatory Box 193.4 and Fig. 193.8),45 bypassing the mechanisms that should nor-
stimulus. However, healthy joints do not contain a significant resident mally regulate these pathways.
population of neutrophils. Monosodium urate crystals have their initial Monosodium urate crystals act as damage-associated molecular patterns
interaction with cells normally resident in the tissue.42,43 Resident mono- (DAMPs) in macrophages.50 This DAMP activity signals macrophage cells to
cytes and macrophages within the synovial membrane play a central role in assemble the NLRP3 inflammasome components, produced in the priming
initiation and driving of the initial inflammatory response to monosodium signal pathway, creating an active NLRP3 inflammasome complex.44
urate crystals within the joint.43 Activation of cells by monosodium urate After NLRP3 inflammasome assembly, caspase 1 is able to cleave the
crystals leads to the production of a diverse array of inflammatory medi- immature proinflammatory interleukins, pro–IL-1β and pro–IL-18, into their
ators, which contribute to massive migration of neutrophils toward the active, inflammation-promoting forms. Interleukin-1β (IL-1β) is among the
monosodium urate crystals that have triggered the inflammatory episode. first inflammatory mediators released after cell exposure to monosodium
Other cells such as synovial fibroblasts, antigen-presenting dendritic cells urate crystals. Release of active IL-1β and IL-18 leads to the rapid recruit-
(DCs), eosinophils, and mast cells may also play a role in the initiation ment of neutrophils to the site of monosodium urate crystal deposition and
phase of the gout flare. full initiation of the acute inflammatory episode.50

F 2 A 2 2, A 2 2 2 A D D 2C ,A2 2AA @DI 1 E A C A ) 2. 3 E A


, 3AD2A , A A 2 D 0 C AD F C DC A ) A C E A ( A C A AE
CHAPTER 193 Etiology and pathogenesis of gout 1707

BOX 193.4 INFLAMMATORY EVENTS IN THE GOUT FLARE


Sequence of cellular events
1. Deposition and release of monosodium urate microcrystals
2. Proinflammatory coating (e.g., IgG, complement)
3. Initial interaction with resident cells: tissue macrophages ± fibroblasts, mast cells, others
4. Activation of membrane signaling molecules (TLR, CD14, TREM)
5. NLRP3 inflammasome activation: mature IL-1β release
6. Release of other cytokines and chemokines
7. Activation of endothelial cell adhesion molecules
8. Emigration, attraction, and activation of neutrophils
9. Phagocytosis of crystals by neutrophils
10. Cross-linking of inflammatory signaling proteins
11. Removal of crystal coating, membrane damage, phagolysosome rupture
12. Enzyme and mediator release
13. Resolution: aggregated NET formation, cytokines TGF-β, MC-R and PPAR-γ ligands, AMPK activation, mature macrophages

Membrane mediators
Fcγ receptors (FcγRIII/CD16) Complement receptors
GPCRs (e.g., chemokine receptors) Integrins
TLRs (TLR-2, TLR-4) CD14

Intracellular signaling
NLRP3 inflammasome activation (caspase 1, PYCARD, NALP)
Tyrosine kinases (Syk, Src, Pyk-2) Lipid kinase PI3K
Mitogen-activated protein kinases (p38, JNK, ERK-1, ERK-2)
Downstream TLR pathway (Myd88, IRAK-1, TRAF-6, IK-κB)
Nuclear factors (NF-κB, AP-1)

Soluble mediators released


Mature IL-1β
Chemokines: CXCL (e.g., IL-8), CCL (e.g., MCP-1)
Complement split products Endothelins
TNF-α IL-6
Kinins Leukotrienes
Metalloproteinases ROS
Reactive nitrogen species (NO) Prostaglandins
S100 proteins (S100A8/9; MRP 8/14) Substance P
TGF-β RANKL

Monosodium urate crystals can activate many cell types and trigger the release of a diverse array of proinflammatory mediators. Multiple pathways have been implicated in different experimental systems and based on
the detection of mediators in patients with gout, and it is likely that some of these mechanisms operate in parallel. The clinical efficacy of the inhibition of prostanoids and IL-1 inhibitors has established their role beyond
doubt. Intervention in vivo with selective inhibitors of other mediators to “prove” their significance in humans has not been reported, and it is more difficult to be certain of the size of their contribution. AMPK, Adenosine
monophosphate–activated protein kinase; AP-1, activator protein 1; CCL, chemokine (C-C motif) ligand; CXCL, C-X-C motif chemokine ligand; ERK, extracellular signal-regulated kinase; IgG, immunoglobulin G; IK-κB,
inhibitor of nuclear factor kappa-B kinase subunit beta; IL, interleukin; IRAK-1, nterleukin-1 receptor-associated kinase 1; JNK, c-Jun N-terminal kinase; MCP-1, monocyte chemotactic factor-1; MC-R, melanocortin
receptors; MRP-4, multidrug resistance protein 4; Myd88, myeloid differentiation primary response 88; NALP, NACHT, LRR, and PYD domains-containing protein; NET, neutrophil extracellular trap; NF-κB, nuclear factor
κB; NLRP3, nucleotide-binding oligomerization domain-like receptor 3; PI3K, phosphatidylinositol-4, 5-bisphosphate 3-kinase; PPAR-γ, peroxisome proliferator-activated receptor γ; PYCARD, apoptosis-associated speck-like
protein containing a CARD; RANKL, receptor activator of nuclear factor-κB ligand; ROS, reactive oxygen species; TGF-β, transforming growth factor-β; TLR, Toll-like receptor; TNF-α, tumor necrosis factor-α; TRAF-6, TNF
receptor associated factor 6; TREM, triggering receptor expressed on myeloid cells.

ACTIVATION OF PHAGOCYTE BY MEMBRANE-ACTIVE CRYSTALS

Phagocytosis by Fusion of vacuolar and Phagolysosome rupture, Cleavage of pro-IL-1, Release of multiple
leukocyte, interaction lysosomal membranes activation of signaling, transcription, proinflammatory
with membrane receptors, inflammasomes and synthesis mediators
and signaling molecules in cytoplasm of mediators
Monosodium
urate
crystallization
Inflammation
Shedding of
preformed
crystals from
tophi

FIG. 193.8 Crystals interact with cell surface receptors and their associated signaling complexes and are phagocytosed by the cell. Rupture of the phagolysosome releases the
crystals into the cytoplasm, allowing interaction with the inflammasome, leading to activation and release of interluekin-1 (IL-1). Signaling protein kinases and promoters for
cytokines and other proinflammatory genes are activated, with the release of a diverse array of inflammatory mediators (see text). Urate and other crystals also interact with
lipid membranes and bind to cytosolic and membrane-bound proteins, acting directly at important steps controlling proinflammatory signaling.

F 2 A 2 2, A 2 2 2 A D D 2C ,A2 2AA @DI 1 E A C A ) 2. 3 E A


, 3AD2A , A A 2 D 0 C AD F C DC A ) A C E A ( A C A AE
1708 SECTION 17 Crystal-Related Arthropathies
microenvironment, nitric oxide can mediate regulatory or direct toxic effects
INFLAMMATORY MEDIATORS INDUCED BY URATE CRYSTALS and can influence neutrophil activation and apoptosis.
Urate crystals
IgG coating Apo-E, B coating NEUTROPHIL ACTIVATION
Resident synovial cells and trafficking monocytes contribute to the initial
burst of proinflammatory mediators important in the initiation of the gout
flare. However, much of the full-blown response observed clinically is medi-
Receptors, ated by neutrophils. Synovial fluid aspirated during gout flares reveals neu-
Stimulates signaling Inhibits trophil counts in a range up to that seen in acute pyogenic septic arthritis.
molecules Neutrophils initially attach to the synovial vascular endothelium through
Responding cell their selectin ligands. Stronger adhesion occurs when the vascular endo-
(macrophage, neutrophil) thelia express integrin molecules in response to chemokines and cytokines
inflammasome activation produced in adjacent tissues. The neutrophils then exit the synovial capillar-
ies and migrate through the synovial matrix to the joint cavity, aided by the
chemotactic gradient. The proinflammatory cytokines prime the neutrophil
S100A8/9 Substance P for an amplified oxidative and degranulation response to crystals.
Endothelins Neutrophils have a shorter half-life (i.e., hours) than any other circu-
lating leukocyte. Unless they are activated, normal neutrophils soon die.
Inflammatory mediators involved in gout, particularly colony-stimulating
Chemokines Lysosomal PGE2 IL-1 Reactive N factors, IL-1, IL-6, chemokines, and prostaglandins, prolong neutrophil sur-
(e.g., CCL2, enzymes LTB4 IL-6 and O metabolites
CXCL8)
vival by inhibiting spontaneous apoptosis.
TNF-!
Neutrophils and macrophages within the synovial fluid avidly engulf
monosodium urate crystals. Lysosomal enzymes then remove the protein
FIG. 193.9 Monosodium urate crystals can stimulate a variety of cells to produce a coating the crystal has acquired in the synovial membrane and cavity. In the
diverse array of inflammatory mediators, depending on the coating of the crystal and uncoated state, monosodium urate crystals have increased reactivity with
the receptiveness of the cell. Many mediators have been implicated in experimental proteins and membranes. Neutrophil activation is marked by activation of
settings, but the dominant mediators appears to be those involved in NLRP3 inflam- phospholipases and by inositol triphosphate production, leading to a cyto-
masome activation. The clinical response of the gout flare to nonsteroidal antiinflam- plasmic calcium flux. Monosodium urate crystals also induce leukotriene
matory drugs and coxibs and to specific interleukin-1 (IL-1) antagonists supports key (LT) production.
roles for prostanoids and for IL-1, respectively. apo, Apolipoprotein; CCL2, chemok- Depending on crystal concentration and coating, monosodium urate
ine (C-C motif) ligand 2; CXCL8, C-X-C Motif Chemokine Ligand 8; IgG, immunoglob- crystals can then lyse the phagolysosomal membrane, spilling the phagocy-
ulin G; LTB4, leukotriene B4; MCP-1, macrophage chemotactic factor 1; N, nitrogen; tosed crystals and the toxic lysosomal contents (proteases, reactive oxygen
O, oxygen; PGE2, prostaglandin E2; TNF-α, tumor necrosis factor-α. species) into the neutrophil cytoplasm (see Fig. 193.8). Phagosome lysis
allows the monosodium urate crystals to interact with the inflammasome
but can also lead to death of the phagocyte; this releases the lysosomal con-
tents together with newly synthesized acute inflammatory mediators.
OTHER INFLAMMATORY MEDIATORS According to the intensity of the response and on the size and number
of joints affected, cytokines, including S100A8/9, IL-1, IL-6, IL-8, MCP-1,
In addition to the release of the proinflammatory cytokines, IL-1β and IL-18, and TNF-α, are produced in sufficient quantity to enter the circulation and
other inflammatory mediators are released when cells interact with mono- stimulate an acute phase response. Patients with gout flares are often sys-
sodium urate crystals, each of which encourages the recruitment of neutro- temically unwell, with fever and leukocytosis, and in some patients display
phils to the site of inflammation. The S100A8/9 protein heterodimer, IL-6, a systemic inflammatory response syndrome easily confused with sepsis (see
granulocyte-macrophage colony stimulating factor (GM-CSF), and tumor Chapter 194).
necrosis factor-α (TNF-α) are among the mediators released within 30 min- The clinical response of gout flares to COX-2 inhibitors and IL-1 antag-
utes of exposure of cells to monosodium urate crystals. onists suggests that prostanoids and IL-1 (see Chapter 195) are prominent
Chemokines are also released in the early stages of inflammation. mediators of the intense acute inflammation characteristic of the gout flare.56
Chemokines of the CXC family are important for attracting and activating Other pain mediators include kinins, neuropeptides such as substance P, and
neutrophils and are major cytokines in acutely inflamed synovial fluid in mast cell products.
gout (Figs. 193.8 and 193.9; see also Box 193.4). Circulating levels of CXCL8
(IL-8) are increased in patients with gout both during flares and during
the intercritical period.51 In addition, CXCR2, CXCL-1 (Gro-α), CXCL-2
TERMINATION OF THE GOUT FLARE
(MIP-2α, Gro-β), and CXCL-3 (MIP-2β, Gro-γ) have been implicated. Even without medical intervention, the gout flare is almost always self-limit-
The chemokine (C-C motif) ligand 2 (CCL2) (monocyte chemotactic ing. The classic flare resolves spontaneously within a week or two. This is an
factor-1 [MCP-1]) aids recruitment of monocytes to sites of inflammation intriguing paradox, given the overlap between gout and other arthropathies
induced by monosodium urate crystals.52 Assigning relative significance to at the level of the molecular mediators involved and the persistence of the
individual chemokines is difficult for several reasons: the chemokine family crystals that initiated the flare.
is large and there is substantial overlap between the various chemokines After ingestion of monosodium urate crystals, neutrophils undergo
binding a given receptor, redundancy in the functions assigned to different NETosis, with formation of neutrophil extracellular traps (NETs).
chemokines and receptors, and incomplete translation between mediator Termination of the gout flare is regulated by formation of aggregated NETs
actions in vitro and in animal models and their functions in human disease. that densely pack monosodium urate crystals and degrade proinflammatory
Another early response to cytokine release is increased expression of cytokines, including IL-1β, TNFα, MCP-1, MIP1α, and IL-6. Neutrophil
E-selectin on nearby vascular endothelial cells. A further mediator involved extracellular trap formation is dependent on reactive oxygen species, which
in experimental crystal-induced inflammation is endothelin-1, a peptide are important in regulation of termination of the inflammation.57
modulator of neutrophil migration produced by vascular endothelium.53 The increase in vascular permeability after acute synovitis has begun
Monosodium urate crystals activate de novo synthesis of inducible cyclo- allows increased entry of antiinflammatory crystal-coating macromolecules
oxygenase (COX-2) and phospholipase-A2, producing the inflammatory from plasma, such as ApoB. Increasing monosodium urate crystal coating
prostanoids prostaglandin-E2 (PGE2) and thromboxane A2 in phagocytes.54 with ApoB and locally produced ApoE and TGF-β can inhibit neutrophil
PGE2 causes vasodilatation, edema, and further leukocyte immigration. The activation.
clinical efficacy of COX inhibition in treating gout flares confirms a signifi- Systemic antiinflammatory mediators include the melanocortins,
cant role for prostanoids in monosodium urate crystal inflammation. products of the hypothalamic–pituitary axis, including adrenocortico-
Monosodium urate crystals also stimulate neutrophils and fibroblast-like tropic hormone (ACTH) and melanocyte stimulating hormone (MSH).
synoviocytes to undergo the respiratory burst and produce reactive oxy- Adrenocorticotropic hormone stimulates the release of corticosteroids from
gen species and reactive nitrogen species (RNS), including nitric oxide the adrenal cortex. Melanocortins also have a direct antiinflammatory effect
(NO).55 The release of nitric oxide involves signaling pathways similar on macrophages in the inflamed joint via macrophage melanocortin recep-
to the proinflammatory cytokines. Depending on its concentration and tors (MCRs).58

F 2 A 2 2, A 2 2 2 A D D 2C ,A2 2AA @DI 1 E A C A ) 2. 3 E A


, 3AD2A , A A 2 D 0 C AD F C DC A ) A C E A ( A C A AE
CHAPTER 193 Etiology and pathogenesis of gout 1709

A number of antiinflammatory mediators have been implicated in the


resolution phase of the gout flare. These include ApoE, TGF-β, IL-10, IL-1ra,
soluble TNF receptors, intracellular cytokine inducible SH2-containing pro-
tein (CIS) and suppressor of cytokine signaling 3 (SOCS 3),59 and inducers
of the nuclear hormone receptor peroxisome proliferator-activated receptor
γ.60 Of particular note is the antiinflammatory cytokine IL-37, which is an
inhibitor of the innate immune system. Mature IL-37 is produced alongside
IL-1β via cleavage by caspase-1; however, IL-37 acts to reduce rather than
promote inflammation. Recent evidence has shown IL-37 can inhibit mono-
sodium urate crystal induced inflammation by downregulating proinflam-
matory mediators, reducing the recruitment of neutrophils and monocytes
to the site of inflammation.61
Resolution of the gout flare may also be aided by the presence of progres-
sively more differentiated macrophages, arising in response to the cytokine
milieu,62 and by removal of neutrophils through apoptosis and phagocyto- FIG. 193.11 Monosodium urate crystals from tophus. Crystals aspirated from a sub-
sis by mature macrophages. However, mature macrophages seem to be as cutaneous tophus. Polarized transmission white (halogen) light; 90-degree extinction
capable as monocytes at initiating responses to monosodium urate crys- filter; 530 nm first-order compensator.
tals.43 Induction of TGF-β through neutrophil cannibalism and the release
of microvesicles (ectosomes) by neutrophils may also play a role in the ter-
mination phase.63,64 Adenosine monophosphate–activated protein kinase During tophus formation, persistent nests of crystals are enveloped by a
(AMPK) is a metabolic biosensor with antiinflammatory activities that plays granuloma-like chronic inflammatory response, with a corona zone of dif-
an important role in the regulation of MSU crystal-associated inflammation. ferentiated macrophages and multinucleate giant cells surrounded in turn
AMPK activators increase macrophage antiinflammatory M2 polarization, by a fibrovascular layer. Both adaptive and innate immune cells are present
inhibit NLRP3 gene expression, and activation of caspase-1 and IL-1β.65 within the tophus, and proinflammatory cytokines, including IL-1 and TNF-
α, are expressed within the corona zone (Fig. 193.10).67 Aggregated neutro-
phil extracellular traps (NETs; discussed above) may also play a role in the
ADVANCED GOUT formation of the tophus.57
In the presence of persistent hyperuricemia, some patients develop advanced Tophi usually develop after years of sustained hyperuricemia, although
gout, with recurrent flares, clinically evident tophi, chronic gouty arthritis, some individuals who present with tophi will have no prior history of gout
and bone erosion. flare. Tophi can be clinically silent for a long time, indicating that the tophus
may represent a physical containment that limits monosodium urate crys-
tal-induced inflammation at the site of crystal deposition.66 Despite this
TOPHUS FORMATION lack of active inflammatory symptoms, the development of tophi can have
The tophus (tophi, plural) is a chronic foreign body granuloma-like struc- serious consequences, limiting joint function and causing bone destruction,
ture containing collections of monosodium urate crystals surrounded by both of which can lead to substantial disabilities.
inflammatory cells and connective tissue (Fig. 193.10 and Fig. 193.11).66
Typically, tophi develop in the first metatarsophalangeal joint, the Achilles
tendon, the olecranon bursa, the ear, and the finger pulps, but they can also
JOINT DAMAGE
develop in other less common sites. Bone erosion occurs in proximity to intraarticular or periarticular tophi
and generally has a radiographic appearance that is distinct from that seen
with the erosions of rheumatoid arthritis. Patients with severe erosive and
tophaceous gout have elevated circulating levels of receptor activator of
nuclear factor-κB ligand (RANKL) and macrophage colony-stimulating fac-
COMPOSITION OF THE GOUTY TOPHUS tor (M-CSF), factors that aid the maturation of bone-resorbing osteoclasts.68
T cells present within the tophus express RANKL and may contribute to
joint damage.69
Monosodium urate crystals also have profound effects on osteoblasts,
Fibrovascular zone
causing reduced cell viability, function, and differentiation.70 These crystals
also reduce the expression of osteoprotegerin, a decoy receptor that inhibits
osteoclastogenesis by preventing the binding of RANKL to its ligand. High
numbers of osteoclasts and infrequent osteoblasts are present at the bone–
tophus interface in patients with advanced gout.68,70 Together, these data
suggest that bone erosion in gout occurs because of uncoupling of osteoclast
and osteoblast function, which in turn promotes bone resorption at sites of
monosodium urate crystal deposition.
Osteocytes are the most abundant cell of bone and play an import-
ant role in regulating bone remodeling. Monosodium urate crystals have
Corona zone direct effects on osteocyte viability and, through interactions with macro-
phages, indirectly promote a shift in osteocyte function that favors bone
resorption and inflammation. These indirect effects can be inhibited by
COX-2 inhibition. Thus the crystals within the tophus may have direct
effects, and the tissue response to the crystals may have indirect effects
on bone cells.71
Cartilage damage, characterized by radiographic joint space narrowing,
is a relatively late manifestation of advanced gout. Histologic studies have
T cell B cell Plasma cell Macrophage shown that monosodium urate crystals are deposited radially in the super-
ficial layers of articular cartilage, and cartilage surfaces in advanced gout
CD68 + MNC Mast cell Neutrophil TRAP + MNC are often described as being diffusely “dusted” with white crystal depos-
its.72 Ultrasound studies have highlighted the close relationship between
monosodium urate crystals and articular cartilage. The “double contour”
FIG. 193.10 The central mass of monosodium urate crystals is surrounded by a corona ultrasound sign can be visualized over the superficial margin of the articular
of cells, including mature macrophages and multinucleated giant cells. Cells of the cartilage in joints of people with asymptomatic hyperuricemia and gout and
adaptive immune response are also present: T and B lymphocytes and plasma cells. is thought to represent monosodium urate crystal deposition.73
MNC, Multinucleated cells; TRAP, tartrate-resistant acid phosphatase. (Adapted Monosodium urate crystals interact with chondrocytes to promote deg-
with permission from Dalbeth N, Pool B, Gamble GD, et al. Cellular characterization radation of the cartilage matrix by inducing nitric oxide generation and
of the gouty tophus: a quantitative analysis. Arthritis Rheum. 2010;62[5]:1549–56.) expression of matrix metalloprotease (MMP)-3 in articular chondrocytes

F 2 A 2 2, A 2 2 2 A D D 2C ,A2 2AA @DI 1 E A C A ) 2. 3 E A


, 3AD2A , A A 2 D 0 C AD F C DC A ) A C E A ( A C A AE
1710 SECTION 17 Crystal-Related Arthropathies
through TLR-2 signaling and upregulation of NF-κB.74 Increased expression the heritability of gout (<10% when stringent criteria are applied).80 This
of other MMPs has also been observed in synovial fibroblasts stimulated suggests that diverse gene variants or a combination of many individually
with monosodium urate crystals75 and in macrophages obtained from syno- “weak” genes contribute to the genetic predisposition to gout. Despite pow-
vial tissue from people with gout and within the tophus.76 erful contemporary epidemiologic and genetic techniques, the relative con-
tributions of genes and environment remain uncertain.
Numerous variants have been identified in the promoter and structural
THE GENETICS OF GOUT regions of genes for inflammatory cytokines, cytokine receptors, and cellu-
A familial diathesis to gout was first noted by Seneca two millennia ago. A lar signaling molecules, including NLRP3 and members of the IL-1 family.
positive family history has been reported in as few as 10% or as many as 80% Expression of one or more of these genes might modulate the inflammatory
of patients with gout. This range reflects differences in the populations stud- response to monosodium urate crystals.88,89 GWASs have also implicated
ied and the definitions used. Genetic influences on hyperuricemia and gout several genes involved in regulation of inflammation.80 Associations between
are, in most instances, multigenic. Contemporary estimates of the heritabil- gout and cytokine gene polymorphisms have not been explored as robustly
ity of serum urate levels have ranged from 40% to 63%, and comparisons of as, for example, the URAT1 and SLC2A9 associations.
contributions to variation in serum urate levels have found a stronger role
for genes over diet or dietary components.77,78
Genes that influence gout could in principle act at any one of the several
RARE GENETIC DISORDERS
“checkpoints” on the path to crystal-induced inflammation (Fig. 193.1). For Gout is not commonly caused by a single-gene disorder. There are, however,
example, they could affect urate production, extrarenal or renal handling of documented cases of rare monogenic disorders resulting in hyperuricemia
uric acid, crystal nucleation and enlargement, the crystal coating, or the cel- and gout. These monogenic disorders provide valuable insight into the bio-
lular and soluble mediator components of the acute response when inflam- logic processes involved in the development of hyperuricemia and gout.
matory cell meets crystal or the amplification of this response. Supporting
the importance of genes influencing uric acid excretion, monozygotic twins Purine metabolism syndromes
show a tighter concordance than dizygotic twins for fractional renal uric Two rare X chromosome–linked disorders, deficiency of the salvage enzyme
acid clearance.79 Genome-wide association studies (GWAS) have identified hypoxanthine-guanine phosphoribosyltransferase (HPRT) and overactivity
common genetic variants within genes involved in urate production, uric of phosphoribosyl pyrophosphate synthetase 1 (PRS-I), provide insight into
acid excretion, control of inflammation, and control of gene expression.80,81 the regulatory processes involved in purine nucleotide synthesis and degra-
Recently, large gene sets related to kidney and liver development, morphol- dation (Fig. 193.6).10
ogy, and function have been identified as being of particular relevance in PRPS1 encodes the PRS-I enzyme, which converts ribose-5-phosphate to
determining serum urate levels.81 phosphoribosyl pyrophosphate (PRPP), the first element in the purine syn-
thesis de novo pathway.90 Rare genetic variants that alter the PRS-I protein
result in super activity of the enzyme, leading to accelerated production of
TRANSPORTER GENES PRPP and consequently overproduction of urate.90
Large, well-powered human genome-wide studies have revealed associ- HPRT is encoded by the HPRT1 gene and catalyzes the conversions of
ations of many genes encoding renal urate transporters with serum urate hypoxanthine to inosine monophosphate (IMP) and guanine to guanosine
concentrations and gout (including SLC2A9, SLC22A12, and ABCG2; along monophosphate (GMP) in the PRPP salvage synthesis pathway. Defects
with SLC2A12, SLC16A9, SLC17A1, SLC17A3, and SLC22A11).80–82 SLC2A9 in this enzyme result in the accumulation of hypoxanthine and guanine,
(GLUT9) has the strongest effect on serum urate levels, and ABCG2 has the resulting in overproduction of urate. Defects in HPRT also result in an
strongest effect on gout risk.80 Variants that reduce protein function for both increase in PRPP, the rate-limiting enzyme in purine synthesis de novo.91
these genes have been identified, with loss-of-function variants in SLC2A9 Overproduction of urate occurs in all individuals with a deficiency of
being linked to renal hypouricemia, and the main ABCG2 variant associated HPRT because of this dysregulation of the purine synthesis de novo path-
with hyperuricemia (Q141K, glutamine to lysine amino acid change) reduc- way. Individuals with severe HPRT-deficiencies also develop neurologic
ing protein function by approximately half.83 SLC2A9 has a stronger effect in conditions.91
women than in men and ABCG2 a stronger effect in men.27,84 The promoter
regions for some of the urate transporter genes contain hormone-responsive Autosomal dominant kidney diseases caused by UMOD
elements that may underlie these effects. pathogenic variants (ADTKD-UMOD)
Similarly, SLC22A12 (URAT1) has a strong effect on serum urate levels. Additional insight into uric acid handling has been provided by studies of
People with loss-of-function variants in the SLC22A12 gene (resulting in patients with autosomal dominant tubulointerstitial kidney disease caused
decreased URAT1 activity) show hypouricemia and hyperuricosuria as well by UMOD pathogenic variants (ADTKD-UMOD), previously described as
as exercise-induced renal functional impairment.85 Consistent with a signif- familial juvenile hyperuricemic nephropathy (FJHN) and medullary cystic
icant role in uric acid excretion, homozygosity for a loss-of-function variant kidney disease (MCKD). These autosomal dominantly inherited disorders
in this gene is the most common cause of renal hypouricemia in Japanese are characterized by early onset of hyperuricemia (with or without gout),
patients.85 This polymorphism is more common in healthy Japanese individ- hypertension, and progressive tubulointerstitial inflammation and fibrosis,
uals than in those with gout, suggesting that it provides protection against culminating in end-stage renal disease, often by age 40 years.
gout.86 Most families with ADTKD-UMOD have genetic variants in uromodulin,
Genetic associations between serum urate level and the PDZK1 gene27 long known as the Tamm-Horsfall urinary glycoprotein.92 Uromodulin plays
further support the importance of the URAT1-PDZ-NPT complex in con- a role in maintaining the integrity of the ascending loop of Henle by forming
trolling net uric acid reabsorption and secretion. a protective gel-like lattice that coats the luminal (urine) side of the tubule.
Dysfunctional uromodulin accumulates in the endoplasmic reticulum.
Defects in the protective lattice alter solute fluxes, reducing Na+ and Cl−
METABOLIC GENES reabsorption. This results in contracted extracellular volume and compensa-
Several well-defined purine metabolic enzyme defects lead to urate over- tory enhancement of sodium-dependent uric acid transport in the proximal
production (see Box 193.2 and below), but these are uncommon. Other tubule.93
metabolic genes have effects that are less direct and less dramatic. There is
an association between serum urate level and GCKR, which encodes glu-
cokinase regulatory protein, a regulator of glucokinase.87 Glucokinase is an
SUMMARY
important enzyme in the glycolytic pathway and acts as a glucose sensor. Gout is a chronic disease of monosodium urate crystal deposition that
GCKR has in turn been linked with metabolic traits associated with gout, typically presents as intermittent episodes of severe joint inflammation.
including insulin resistance and hyperlipidemia. Other implicated genes are Dysregulation of the pathways involved in regulating urate production and
involved in lipid or sugar metabolism and in the production of purine pre- excretion, leading to hyperuricemia, is essential in gout. However, there
cursors through the pentose phosphate pathway.80–82 are a number of other important “checkpoints” that determine whether
an individual will develop clinically evident gout, including monosodium
GENES INFLUENCING CRYSTAL FORMATION AND CRYSTAL- urate crystal formation, and the onset of an acute inflammatory response
(Fig. 193.1). The development of advanced gout represents chronic inflam-
TRIGGERED INFLAMMATION mation and tophus formation. Genetics, environmental factors, other clin-
Many people with hyperuricemia do not develop gout. Moreover, the genes ical conditions, and medications are all able to influence the pathways
currently known to influence urate level account for a small proportion of involved in reaching these “checkpoints.”

F 2 A 2 2, A 2 2 2 A D D 2C ,A2 2AA @DI 1 E A C A ) 2. 3 E A


, 3AD2A , A A 2 D 0 C AD F C DC A ) A C E A ( A C A AE
CHAPTER 193 Etiology and pathogenesis of gout 1711

ACKNOWLEDGMENT 33. Muehleman C, Li J, Aigner T, et al. Association between crystals and cartilage degeneration
in the ankle. J Rheumatol. 2008;35:1108–1117.
The authors wish to acknowledge Professor Michael A. Becker and Dr. Lachy 34. Katz WA, Schubert M. The interaction of monosodium urate with connective tissue compo-
nents. J Clin Invest. 1970;49:1783–1789.
McLean for their contributions to previous versions of this chapter.
35. Laurent TC. Solubility of sodium urate in the presence of chondroitin-4-sulphate. Nature.
1964;202:1334.
REFERENCES 36. Burt HM, Dutt YC. Growth of monosodium urate monohydrate crystals: effect of cartilage
and synovial fluid components on in vitro growth rates. Ann Rheum Dis. 1986;45:858–864.
1. Wu XW, Muzny DM, Lee CC, et al. Two independent mutational events in the loss of urate 37. Chhana A, Pool B, Wei Y, et al. Human cartilage homogenates influence the crystallization
oxidase during hominoid evolution. J Mol Evol. 1992;34:78–84. of monosodium urate and inflammatory response to monosodium urate crystals: a potential
2. Hershfield MS, Roberts LJ, Ganson NJ, et al. Treating gout with pegloticase, a PEGylated link between osteoarthritis and gout. Arthritis Rheumatol. 2019;71:2090–2099.
urate oxidase, provides insight into the importance of uric acid as an antioxidant in vivo. 38. De Miguel E, Puig JG, Castillo C, et al. Diagnosis of gout in patients with asymptomatic
Proc Natl Acad Sci USA. 2010;107:14351–14356. hyperuricaemia: a pilot ultrasound study. Ann Rheum Dis. 2012;71:157–158.
3. Shi Y, Evans JE, Rock KL. Molecular identification of a danger signal that alerts the immune 39. Terkeltaub R, Martin J, Curtiss LK, Ginsberg MH. Apolipoprotein B mediates the capacity
system to dying cells. Nature. 2003;425:516–521. of low density lipoprotein to suppress neutrophil stimulation by particulates. J Biol Chem.
4. Watanabe S, Kang DH, Feng L, et al. Uric acid, hominoid evolution, and the pathogenesis of 1986;261:15662–15667.
salt-sensitivity. Hypertension. 2002;40:355–360. 40. Dalbeth N, Haskard DO. Mechanisms of inflammation in gout. Rheumatology.
5. Campion EW, Glynn RJ, DeLabry LO. Asymptomatic hyperuricemia. Risks and conse- 2005;44:1090–1096.
quences in the Normative Aging Study. Am J Med. 1987;82:421–426. 41. Gordon TP, Bertouch JV, Walsh BR, Brooks PM. Monosodium urate crystals in asymptomatic
6. Zhu Y, Pandya BJ, Choi HK. Prevalence of gout and hyperuricemia in the US general popu- knee joints. J Rheumatol. 1982;9:967–969.
lation: the National Health and Nutrition Examination Survey 2007-2008. Arthritis Rheum. 42. Chen C-J, Shi Y, Hearn A, et al. MyD88-dependent IL-1 receptor signaling is essential for
2011;63:3136–3141. gouty inflammation stimulated by monosodium urate crystals. [see comment]. J Clin Invest.
7. Wyngaarden JB, Kelley WN. Gout and hyperuricemia. New York: Grune & Stratton; 1976. 2006;116:2262–2271.
1-503 and references therein. 43. Martin WJ, Walton M, Harper J. Resident macrophages initiating and driving inflammation
8. Munan L, Kelly A, Petitclerc C. Serum urate levels between ages 10 and 14: changes in sex in a monosodium urate monohydrate crystal-induced murine peritoneal model of acute
trends. J Lab Clin Med. 1977;90:990–996. gout. Arthritis Rheum. 2009;60:281–289.
9. Antón FM, Puig JG, Ramos T, et al. Sex differences in uric acid metabolism in adults: 44. Swanson KV, Deng M, Ting JP. The NLRP3 inflammasome: molecular activation and regula-
evidence for a lack of influence of estradiol-17 beta (E2) on the renal handling of urate. tion to therapeutics. Nature Reviews Immunology. 2019;19:477–489.
Metabolism. 1986;35:343–348. 45. So AK, Martinon F. Inflammation in gout: mechanisms and therapeutic targets. Nature
10. Becker MA. Hyperuricemia and gout. In: Scriver CR, ed. The metabolic and molecular bases Reviews Rheumatology. 2017 Nov;13:639.
of inherited disease. New York: McGraw-Hill; 2001:2513–2535. 46. Joosten LA, Netea MG, Mylona E, et al. Engagement of fatty acids with Toll-like receptor 2
11. Sarawate CA, Patel PA, Schumacher HR, et al. Serum urate levels and gout flares: analysis drives interleukin-1beta production via the ASC/caspase 1 pathway in monosodium urate
from managed care data. J Clin Rheumatol. 2006;12:61–65. monohydrate crystal-induced gouty arthritis. Arthritis Rheum. 2010;62:3237–3248.
12. Woodward OM, Köttgen A, Coresh J, et al. Identification of a urate transporter, ABCG2, 47. Liu-Bryan R, Scott P, Sydlaske A, et al. Innate immunity conferred by Toll-like receptors
with a common functional polymorphism causing gout. Proc Natl Acad Sci USA. 2 and 4 and myeloid differentiation factor 88 expression is pivotal to monosodium urate
2009;106:10338–10342. monohydrate crystal-induced inflammation. Arthritis Rheum. 2005;52:2936–2946.
13. Ichida K, Matsuo H, Takada T, et al. Decreased extra-renal urate excretion is a common 48. Martinon F, Pétrilli V, Mayor A, et al. Gout-associated uric acid crystals activate the NALP3
cause of hyperuricemia. Nat Commun. 2012;3:764. inflammasome. Nature. 2006;440:237–241.
14. Perez Ruiz F, Calabozo M, Erauskin GG, et al. Renal underexcretion of uric acid is present 49. Schlesinger N, Kay JC. Gouty Inflammation. Textbook of Autoinflammation. Cham: Springer;
in patients with apparent high urinary uric acid output. Arthritis Rheum. 2002;47:610–613. 2019:635–645.
15. Simkin PA. New standards for uric acid excretion and evidence for an inducible transporter. 50. Ryckman C, McColl SR, Vandal K, et al. Role of S100A8 and S100A9 in neutrophil recruit-
Arthritis Rheum. 2003;49:735–736. ment in response to monosodium urate monohydrate crystals in the air-pouch model of
16. Lipkowitz MS, Leal-Pinto E, Rappoport JZ, et al. Functional reconstitution, membrane acute gouty arthritis. Arthritis Rheum. 2003;48:2310–2320.
targeting, genomic structure, and chromosomal localization of a human urate transporter. 51. Kienhorst LB, van Lochem E, Kievit W, et al. Gout is a chronic inflammatory disease in
J Clin Invest. 2001;107:1103–1115. which high levels of interleukin-8 (CXCL8), myeloid-related protein 8/myeloid-related pro-
17. Anzai N, Ichida K, Jutabha P, et al. Plasma urate level is directly regulated by a voltage-driven tein 14 complex, and an altered proteome are associated with diabetes mellitus and cardio-
urate efflux transporter URATv1 (SLC2A9) in humans. J Biol Chem. 2008;283:26834–26838. vascular disease. Arthritis Rheumatol. 2015;67:3303–3313.
18. Caulfield MJ, Munroe PB, O’Neill D, et al. SLC2A9 is a high-capacity urate transporter in 52. Matsukawa A, Miyazaki S, Maeda T, et al. Production and regulation of monocyte chemo-
humans. PLoS Med. 2008;5:e197. attractant protein-1 in lipopolysaccharide- or monosodium urate crystal-induced arthritis
19. Matsuo H, Chiba T, Nagamori S, et al. Mutations in glucose transporter 9 gene SLC2A9 in rabbits: roles of tumor necrosis factor alpha, interleukin-1, and interleukin-8. Lab Invest.
cause renal hypouricemia. [erratum appears in Am J Hum Genet. 2008;83(6):795]. Am J 1998;78:973–985.
Hum Genet. 2008;83:744–751. 53. Getting SJ, Di Filippo C, Lam CW, et al. Investigation into the potential anti-inflammatory
20. Preitner F, Bonny O, Laverrière A, et al. Glut9 is a major regulator of urate homeostasis and effects of endothelin antagonists in a murine model of experimental monosodium urate
its genetic inactivation induces hyperuricosuria and urate nephropathy. Proc Natl Acad Sci peritonitis. J Pharmacol Exp Ther. 2004;310:90–97.
USA. 2009;106:15501–15506. 54. Nalbant S, Chen LX, Sieck MS, et al. Prophylactic effect of highly selective COX-2 inhibi-
21. Enomoto A, Kimura H, Chairoungdua A, et al. Molecular identification of a renal urate tion in acute monosodium urate crystal induced inflammation in the rat subcutaneous air
anion exchanger that regulates blood urate levels. Nature. 2002;417:447–452. pouch. Journal of Rheumatology. 2005;32:1762–1764.
22. Sweet DH, Chan LM, Walden R, et al. Organic anion transporter 3 (Slc22a8) is a dicar- 55. Zamudio-Cuevas Y, Martínez-Flores K, Fernández-Torres J, et al. Monosodium urate crys-
boxylate exchanger indirectly coupled to the Na+ gradient. Am J Physiol Renal Physiol. tals induce oxidative stress in human synoviocytes. Arthritis Res Ther. 2016;18:117.
2003;284:F763–F769. 56. Torres R, MacDonald L, Croll SD, et al. Hyperalgesia, synovitis and multiple biomarkers of
23. Eraly SA, Vallon V, Rieg T, et al. Multiple organic anion transporters contribute to net renal inflammation are suppressed by interleukin 1 inhibition in a novel animal model of gouty
excretion of uric acid. Physiol Genomics. 2008;33:180–192. arthritis. Ann Rheum Dis. 2009;68:1602–1608.
24. Urano W, Taniguchi A, Anzai N, et al. Sodium-dependent phosphate cotransporter type 1 57. Schauer C, Janko C, Munoz LE, et al. Aggregated neutrophil extracellular traps limit inflam-
sequence polymorphisms in male patients with gout. Ann Rheum Dis. 2010;69:1232–1234. mation by degrading cytokines and chemokines. Nat Med. 2014;20:511–517.
25. Van Aubel RA, Smeets PH, van den Heuvel JJ, Russel FG. Human organic anion trans- 58. Getting SJ, Christian HC, Flower RJ, Perretti M. Activation of melanocortin type 3 recep-
porter MRP4 (ABCC4) is an efflux pump for the purine end metabolite urate with multiple tor as a molecular mechanism for adrenocorticotropic hormone efficacy in gouty arthritis.
allosteric substrate binding sites. Am J Physiol Renal Physiol. 2005;288:F327–F333. Arthritis Rheum. 2002;46:2765–2775.
26. Hosomi A, Nakanishi T, Fujita T, Tamai I. Extra-renal elimination of uric acid via intestinal 59. Chen YH, Hsieh SC, Chen WY, et al. Spontaneous resolution of acute gouty arthritis is
efflux transporter BCRP/ABCG2. PloS ONE. 2012;7:e30456. associated with rapid induction of the anti-inflammatory factors TGFbeta1, IL-10 and sol-
27. Kolz M, Johnson T, Sanna S, et al. Meta-analysis of 28, 141 individuals identifies com- uble TNF receptors and the intracellular cytokine negative regulators CIS and SOCS3. Ann
mon variants within five new loci that influence uric acid concentrations. PLoS Genet. Rheum Dis. 2011;70:1655–1663.
2009;5:e1000504. 60. Akahoshi T, Namai R, Murakami Y, et al. Rapid induction of peroxisome proliferator-activated
28. McGill NW, Dieppe PA. The effect of biological crystals and human serum on the rate receptor gamma expression in human monocytes by monosodium urate monohydrate crys-
of formation of crystals of monosodium urate monohydrate in vitro. Br J Rheumatol. tals. Arthritis Rheum. 2003;48:231–239.
1991;30:107–111. 61. Liu L, Xue Y, Zhu Y, et al. Interleukin 37 limits monosodium urate crystal-induced innate
29. Kam M, Perl-Treves D, Caspi D, Addadi L. Antibodies against crystals. FASEB J. immune responses in human and murine models of gout. Arthritis Res Ther. 2016;18:268.
1992;6:2608–2613. 62. Landis RC, Yagnik DR, Florey O, et al. Safe disposal of inflammatory monosodium urate
30. Kanevets U, Sharma K, Dresser K, Shi Y. A role of IgM antibodies in monosodium urate monohydrate crystals by differentiated macrophages. Arthritis Rheum. 2002;46:3026–3033.
crystal formation and associated adjuvanticity. J Immunol. 2009;182:1912–1918. 63. Steiger S, Harper JL. Neutrophil cannibalism triggers transforming growth factor beta1
31. Dalbeth N, Doyle A, Boyer L, et al. Development of a computed tomography method of scor- production and self regulation of neutrophil inflammatory function in monosodium urate
ing bone erosion in patients with gout: validation and clinical implications. Rheumatology monohydrate crystal-induced inflammation in mice. Arthritis Rheum. 2013;65:815–823.
(Oxford). 2011;50:410–416. 64. Cumpelik A, Ankli B, Zecher D, Schifferli JA. Neutrophil microvesicles resolve gout
32. Roddy E, Zhang W, Doherty M. Are joints affected by gout also affected by osteoarthritis? by inhibiting C5a-mediated priming of the inflammasome. Ann Rheum Dis. 2016;75:
Ann Rheum Dis. 2007;66:1374–1377. 1236–1245.

F 2 A 2 2, A 2 2 2 A D D 2C ,A2 2AA @DI 1 E A C A ) 2. 3 E A


, 3AD2A , A A 2 D 0 C AD F C DC A ) A C E A ( A C A AE
1712 SECTION 17 Crystal-Related Arthropathies
65. Wang Y, Viollet B, Terkeltaub R, Liu-Bryan R. AMP-activated protein kinase suppresses urate 79. Emmerson BT, Nagel SL, Duffy DL, Martin NG. Genetic control of the renal clearance of
crystal-induced inflammation and transduces colchicine effects in macrophages. Ann Rheum urate: a study of twins. Ann Rheum Dis. 1992;51:375–377.
Dis. 2016;75:286–294. 80. Kottgen A, Albrecht E, Teumer A, et al. Genome-wide association analyses identify 18 new
66. Chhana A, Dalbeth N. The gouty tophus: a review. Curr Rheumatol Rep. 2015;17:19. loci associated with serum urate concentrations. Nat Genet. 2013;45:145–154.
67. Dalbeth N, Pool B, Gamble GD, et al. Cellular characterization of the gouty tophus: a quan- 81. Tin A, Marten J, Kuhns VL, et al. Target genes, variants, tissues and transcriptional pathways
titative analysis. Arthritis Rheum. 2010;62:1549–1556. influencing human serum urate levels. Nat Genet. 2019;2:1–6.
68. Dalbeth N, Smith T, Nicolson B, et al. Enhanced osteoclastogenesis in patients with topha- 82. Matsuo H, Yamamoto K, Nakaoka H, et al. Genome-wide association study of clinically
ceous gout: urate crystals promote osteoclast development through interactions with stro- defined gout identifies multiple risk loci and its association with clinical subtypes. Ann
mal cells. Arthritis Rheum. 2008;58:1854–1865. Rheum Dis. 2016;75:652–659.
69. Lee SJ, Nam KI, Jin HM, et al. Bone destruction by receptor activator of nuclear factor kap- 83. Matsuo H, Ichida K, Takada T, et al. Common dysfunctional variants in ABCG2 are a major
paB ligand-expressing T cells in chronic gouty arthritis. Arthritis Res Ther. 2011;13:R164. cause of early-onset gout. Sci Rep. 2013;3:2014.
70. Chhana A, Callon KE, Pool B, et al. Monosodium urate monohydrate crystals inhibit osteo- 84. Döring A, Gieger C, Mehta D, et al. SLC2A9 influences uric acid concentrations with pro-
blast viability and function: implications for development of bone erosion in gout. Ann nounced sex-specific effects. Nat Genet. 2008;40:430–436.
Rheum Dis. 2011;70:1684–1691. 85. Ichida K, Hosoyamada M, Hisatome I, et al. Clinical and molecular analysis of patients with
71. Chhana A, Pool B, Callon KE, et al. Monosodium urate crystals reduce osteocyte viability renal hypouricemia in Japan-influence of URAT1 gene on urinary urate excretion. J Am Soc
and indirectly promote a shift in osteocyte function towards a proinflammatory and prore- Nephrol. 2004;15:164–173.
sorptive state. Arthritis Res Ther. 2018;20:208. 86. Taniguchi A, Urano W, Yamanaka M, et al. A common mutation in an organic anion trans-
72. Levin MH, Lichtenstein L, Scott HW. Pathologic changes in gout; survey of eleven necrop- porter gene, SLC22A12, is a suppressing factor for the development of gout. Arthritis
sied cases. Am J Pathol. 1956;32:871–895. Rheum. 2005;52:2576–2577.
73. Grassi W, Meenagh G, Pascual E, Filippucci E. “Crystal clear”-sonographic assess- 87. van der Harst P, Bakker SJ, de Boer RA, et al. Replication of the five novel loci for uric acid
ment of gout and calcium pyrophosphate deposition disease. Semin Arthritis Rheum. concentrations and potential mediating mechanisms. Hum Mol Genet. 2010;19:387–395.
2006;36:197–202. 88. McKinney C, Stamp LK, Dalbeth N, et al. Multiplicative interaction of functional inflam-
74. Liu-Bryan R, Pritzker K, Firestein GS, Terkeltaub R. TLR2 signaling in chondrocytes drives masome genetic variants in determining the risk of gout. Arthritis Res Ther. 2015;17:288.
calcium pyrophosphate dihydrate and monosodium urate crystal-induced nitric oxide gen- 89. Qing YF, Zhou JG, Zhang QB, et al. Association of TLR4 Gene rs2149356 polymorphism
eration. J Immunol. 2005;174:5016–5023. with primary gouty arthritis in a case-control study. PLoS ONE. 2013;8:e64845.
75. McMillan RM, Vater CA, Hasselbacher P, et al. Induction of collagenase and prostaglan- 90. de Brouwer AP, van Bokhoven H, Nabuurs SB, et al. PRPS1 mutations: four distinct syn-
din synthesis in synovial fibroblasts treated with monosodium urate crystals. J Pharm dromes and potential treatment. Am J Hum Genet. 2010;86:506–518.
Pharmacol. 1981;33:382–383. 91. Torres RJ, Puig JG. Hypoxanthine-guanine phosophoribosyltransferase (HPRT) deficiency:
76. Schweyer S, Hemmerlein B, Radzun HJ, Fayyazi A. Continuous recruitment, co-expression Lesch-Nyhan syndrome. Orphanet J Rare Dis. 2007;2:48.
of tumour necrosis factor-alpha and matrix metalloproteinases, and apoptosis of macro- 92. Hart TC, Gorry MC, Hart PS, et al. Mutations of the UMOD gene are responsible for medul-
phages in gout tophi. Virchows Arch. 2000;437:534–539. lary cystic kidney disease 2 and familial juvenile hyperuricaemic nephropathy. J Med Genet.
77. Kuo CF, Grainge M, See LC, et al. Familial aggregation and heritability of gout in taiwan: a 2002;39:882–892.
nationwide population study. Arthritis Rheum. 2012;64:S355. 93. Dahan K, Devuyst O, Smaers M, et al. A cluster of mutations in the UMOD gene causes
78. Major TJ, Topless RK, Dalbeth N, Merriman TR. Evaluation of the diet wide contribution to familial juvenile hyperuricemic nephropathy with abnormal expression of uromodulin.
serum urate levels: meta-analysis of population based cohorts. BMJ. 2018;363:k3951. J Am Soc Nephrol. 2003;14:2883–2893.

F 2 A 2 2, A 2 2 2 A D D 2C ,A2 2AA @DI 1 E A C A ) 2. 3 E A


, 3AD2A , A A 2 D 0 C AD F C DC A ) A C E A ( A C A AE

You might also like