You are on page 1of 13

International Journal of Toxicology

1-13
Kidney Pathophysiology, Toxicology, and ª The Author(s) 2019
Article reuse guidelines:
sagepub.com/journals-permissions
Drug-Induced Injury in Drug Development DOI: 10.1177/1091581819831701
journals.sagepub.com/home/ijt

Zaher A. Radi1

Abstract
Anatomically, the kidneys are paired, bean-shaped (in most mammals), excretory organs that lie in the retroperitoneum. High
blood flow to the kidneys, together with high oxygen consumption, makes them more vulnerable to exposure, via the circulation,
and subsequent injury related to high concentrations of xenobiotics and chemicals. In preclinical drug development and safety
assessment of new investigational drugs, changes in kidney structure and/or function following drug administration in experi-
mental laboratory animals need to be put in context with interspecies differences in kidney functional anatomy, physiology,
spontaneous pathologies, and toxicopathological responses to injury. In addition, translation to human relevance to avoid pre-
mature drug termination from development is vital. Thus, detection and characterization of kidney toxicity in preclinical species
and human relevance will depend on the preclinical safety testing strategy and collective weight-of-evidence approach including
new investigational drug mechanism of action (MOA), preclinical and clinical interspecies differences, and MOA relevance to
humans. This review describes kidney macroscopic and microscopic functional anatomy, physiology, pathophysiology, toxicology,
and drug-induced kidney toxicities in safety risk assessment and drug development.

Keywords
kidney, pathophysiology, toxicology, drug-induced injury, drug development

Introduction emerging technologies bridging the preclinical to clinical gap.


Specifically, 4 talks highlighted: (1) basic kidney histologic
Understanding the science of kidney injury with respect to anatomy, physiology, and toxicologic pathology; (2) mechan-
functional anatomy, pathophysiology, mechanisms and bio- isms of drug-induced kidney toxicity and their interpretations;
markers of nephrotoxicity, and alternative models that allow (3) translational biomarkers for early detection of kidney
human translation of preclinical data is very critical for phar- injury; and (4) application of kidney-on-a-chip microphysiolo-
maceutical drug development, chemical hazard risk assess- gical system for kidney safety and regulatory assessment.
ment, and consumer health industries. In this review, the
basic anatomy of the normal kidney tubular, interstitial, vascu-
lar, and glomerular structures and their function are discussed. Kidney Macroscopic and
In addition, the basic general pathological terms applicable to Microscopic Anatomy
kidney commonly used in toxicologic pathology are presented. Kidney Macroscopic Anatomy
Examples of toxicologically relevant interspecies physiologic
and developmental differences are discussed. In addition, some The renal system is comprised of 2 kidneys, 2 ureters, a urinary
examples of common drug-induced tubular, interstitial, vascu- bladder, and a urethra. The kidneys are paired, bean-shaped (in
lar, and glomerular pathologies and toxicities encountered in most mammals), excretory organs that lie in the retroperito-
drug development along with preclinical to clinical translation neum and are situated in the posterior part of the abdomen
and human relevance of some of these toxicities are presented. on each side of the vertebral column. The right kidney is lower
This review is based on a symposium entitled “The Brilliant than the left kidney. Kidneys weight varies with body surface
Kidney in Drug Development: From Basics to Chips” at the
Annual Meeting of the American College of Toxicology (West 1
Pfizer Worldwide Research and Development, Drug Safety R&D, Cambridge,
Palm Beach, Florida, 2018). The objective of this symposium MA, USA
was to provide current state, innovation, and anticipated reg-
ulatory changes as it relates to kidney efficacy and safety risk Corresponding Author:
Zaher A. Radi, DVM, MBA, PhD, DABT, DACVP, Pfizer Worldwide Research,
assessment. The symposium covered basic, yet relevant kidney Development, and Medical, Drug Safety Research Development, One Portland
general physiology and drug-induced pathologies and case Street, Cambridge, MA 02139, USA.
examples to illustrate recent advances in biomarkers and Email: zaher.radi@Pfizer.com
2 International Journal of Toxicology XX(X)

area, age, and sex, and both kidneys weigh approximately


0.51% to 1.08% (a mean of 0.65%) of the body weight.1,2
Despite this low weight relative to body weight, the kidneys
receive 25% of the cardiac output, with 85% of renal blood
flow in the cortex, 14% in the outer medulla, and only 1% in the
inner medulla.1,2 Such significant kidney microanatomic dif-
ferences in perfusion and blood flow and oxygen consumption
makes the kidneys more vulnerable to exposure, via the circu-
lation, and subsequent injury related to high concentrations of
xenobiotics and chemicals.2 The main renal artery, which ori-
ginates from the abdominal aorta, branches between kidney
parenchymal lobes to form the interlobar arteries and these
arteries extend to the corticomedullary junction to form the
arcuate arteries (ie, these arteries arch between the cortex and
the medulla), which run parallel to the capsule. Interlobular
arteries give rise to afferent arterioles that give rise to glomer-
ular capillaries (ie, capillary loops). When these capillaries exit
the glomerulus, they exit as efferent arterioles that further
branch to form peritubular (ie, interstitial) capillaries. Efferent
arterioles extend into the medulla as vasa recta and supply the
outer and inner medullae. The vessels of the venous system run
parallel to the arterial vessels and form the interlobular vein, Figure 1. Microscopic anatomical regions of the rat kidney. Note
arcuate vein, interlobar vein, and renal vein. The vasa recta and inner stripe of the outer medulla (ISOM), outer stripe of the outer
peritubular capillaries drain into interlobular veins3; the veins medulla (OSOM), very well-developed OSOM, and unipapillary, single
leave the kidneys as renal veins, which empty into the inferior renal pyramid. Hematoxylin and eosin stain. Source: Reprinted from
Khan, Hard, and Radi (2012), ©2012.
vena cava. Lymphatics run parallel only to the cortical
vasculature.3
Each kidney is surrounded by a fibrous capsule, which is nephrons. The nephron consists of glomeruli, renal tubules
smooth and can be easily removed at autopsy in healthy kid- (proximal tubules, loop of Henle, distal tubules, connecting
neys. Surrounding the renal capsule is a thick layer of perineph- segment, collecting ducts), interstitium, and juxtaglomerular
ric fat to provide protection from trauma. Macroscopically on apparatus (JGA). The initial plasma filtering portion of the
cut surface at autopsy, the kidney parenchyma has 4 major nephron is the glomerulus, which is contained within Bowman
anatomical regions: (1) the superficial cortex (a pale outer capsule. Microscopically, the glomerulus is composed of capil-
region) that contains renal corpuscles, (2) the deep medulla lary network lined by a thin layer of fenestrated endothelial
(a darker inner region) with conical medullary pyramids, (3) cells, glomerular basement membrane (GBM), a central region
renal papilla, and (4) renal pelvis. The medulla is further sub- composed of mesangial cells and matrix, visceral (inner)
divided into the outer medulla and the inner medulla. The outer epithelial cells (podocytes), and parietal (outer) epithelial cells.
medulla is divided into the outer stripe of the outer medulla and Afferent arterioles supply blood to the glomerulus, while effer-
the inner stripe of the outer medulla (Figure 1). The outer stripe ent arterioles exit the glomerulus. The glomerular filtration
contains parts of the proximal tubule (thick descending limbs barrier (size, shape, and charge selective) has 3 layers: the
of Henle’s loop), straight parts of the distal tubule (thick fenestrated endothelium, a negatively charged GBM, and the
ascending limbs), and collecting ducts. The inner stripe com- podocytes. During the filtration process through glomerular
prises descending thin limbs, ascending thick limbs, and col- capillaries, it is important not to overload such process as this
lecting ducts. The inner medulla is established by thin limbs might lead to glomerular lesions and proteinuria such as the
(descending and ascending) and collecting ducts.4 In the lower case with glomerular calcification that might be seen after
urinary tract, ureters transport urine from the kidneys to the administration of dibasic sodium phosphate solution in rats.5
urinary bladder. The urinary bladder serves as a temporary The glomerulus opens into the proximal convoluted tubules,
storage reservoir for urine before the urethra transports urine and in mature male mice, the proximal convoluted tubulars can
out of the body. The urethra of females is short and is longer in extend into Bowman capsule.2 The JGA is located at the vas-
males. The structure and function of the lower urinary tract is cular pole of the glomerulus. The components of the JGA
generally similar in all mammals.1,2 include the macula densa of the thick ascending limb, afferent
and efferent arterioles, granular cells of the afferent arteriole
that produce renin and angiotensin II (AII), and the extraglo-
Kidney Microscopic Anatomy merular mesangial cells. The JGA is important in tubuloglo-
The urine-producing functional and structural units of the kid- merular feedback control of renin secretion produced by the
ney that are present in the cortex and medulla are called granular cells.2
Radi 3

Cortical proximal convoluted tubular epithelial cells are cover the luminal surface of the cells lining the TAL. The
cuboidal cells with well-developed, dense surface microvilli collecting duct system (cortical collecting ducts, the outer
(brush border).3 The first segment of the proximal tubule is medullary collecting ducts, and the inner medullary collecting
convoluted and is called pars convolute, and the subsequent ducts) extends from the connecting segment in the cortex
part that passes down toward the medulla to become the des- through the outer and the inner medullae to the tip of the
cending limb of the loop of Henle is straight and is called pars papilla. The 2 cell types in this system are the intercalated
recta. Thus, the segments (S) of proximal tubules can be sub- cell (a dark cell) and the principal cell with amphophilic or
divided into the S1 and S2 (convoluted) and the S3 (pars recta clear cytoplasm. In the inner medulla, the intercalated cell
or straight) in experimental laboratory animals (rats, mice, and is absent. In the papilla, collecting ducts form the ducts of
monkeys).2 These segments have different energy and oxygen Bellini, which empty into the pelvis.2
requirements and can be distinguished using various markers
via immunohistochemical techniques. The first part of the
proximal convoluted tubules, S1 segment, is short, connects
Kidney Interspecies Macroscopic and Microscopic
with the glomerular filtration space, contains numerous mito-
chondria with highest oxidative metabolic rate, and its brush
Anatomical Differences
borders and phagosomal system are well developed as this The interspecies and sex differences in kidney macroscopic
segment is involved in reabsorption and degradation of macro- and microscopic anatomy among experimental laboratory
molecules. The S2 segment represents most of the proximal animals used in preclinical safety testing versus humans
tubules, has a shorter brush border with fewer mitochondria need to be considered when evaluating and interpreting test
but has more lysosomal bodies, and is the primary site for article–related kidney changes and pathologies. For exam-
low-molecular-weight protein resorption. The S3 segment has ple, the kidney medulla in humans is multipapillary (multi-
a tall brush border and peroxisomes and is a primary site for lobular) because it is divided into 8 to 18 conical masses
cytochrome P450 metabolizing enzymes. This is important as (pyramids; lobes). Similarly, minipigs have multilobular
some drugs might cause tubular hypertrophy in the S3 segment kidney. Rat, mice, dogs, and cynomolgus monkeys have a
that is related to peroxisome proliferation. Mixed-function oxi- unipapillary, single renal pyramid (Figure 1; Table 1).2,7 In
dase activity occurs primarily in pars recta in the rat. The addition, simple (ie, has vascular bundles consisting only of
cortical distal convoluted tubules have very few microvilli and descending and ascending vasa recta) renal medulla anato-
connect the nephron with the collecting duct. In rodents, the mical structure is present in most mammals, including
distal convoluted tubules contain Tamm-Horsfall protein, a humans. However, a complex (ie, descending thin limbs
mucoprotein.2 of short loops that are integrated into vascular bundles)
The spaces among various kidney microscopic anatomic renal medulla is present in mammals (eg, rat and mouse)
structures (the intertubular, extraglomerular, extravascular with a high urine concentrating ability with thickest renal
spaces) are defined as the interstitium. These spaces in the medulla 2,4 (Table 1). Therefore, while humans have an
cortex and medulla (ie, cortical and medullary interstitium) osmolality approximately up to 300 mOsm/kg, rats and mice
contain different cellular constituents, extracellular matrix, and osmolality can be up to 4,300 mOsm/kg and this would, for
interstitial fluid. The interstitial space accounts for approxi- example, affect drug-related kidney crystalluria formation, a
mately 7% to 8% of the total parenchymal volume in the cortex finding in rodents that is not relevant to humans. Mice have
and is thicker and accounts for approximately 29% to 40% in long loops of Henle and associated vasa recta in the
the inner medulla. The cells in the interstitium include dendritic medulla. In rats, the outer stripe is very well developed and
cells (DCs), macrophages, lymphocytes, lymphatic endothelial makes up about one-third of the outer medulla (Figure 1)7,
cells, and fibroblasts. The cells in the interstitium of the papilla very thin in humans and nonhuman primates, and is almost
include stellate cells, monocytes, and pericytes. The proportion completely absent in dogs.2,4,7 In dogs, all nephrons are of
of interstitium in the inner medulla increases with age and the long-looped variety, whereas in rats and mouse, long-
ischemia. The papillary interstitium is rich in mucopolysac- looped nephrons account for 28% of the total nephrons.2
charides matrix.2,6 Normal sexual dimorphism can be noted in both male and
The inner stripe of the outer zone of the medulla consists of female mice (eg, larger kidney corpuscles noted in males
Henle loops, thick ascending tubules (thick ascending seg- than females). The epithelium lining Bowman capsule in the
ment or straight portion of the distal tubule), and collecting kidney of male mice is cuboidal, while in females the
ducts.2 The inner zone of the medulla consists of Henle loops epithelium is simple squamous. This should not be confused
and collecting ducts. The thick ascending limb of the loop of with drug-induced epithelial hyperplasia. Also, unlike other
Henle (TAL) is one of the 3 structural segments of the distal species, mice excrete large amounts of protein in the urine
tubules. The TAL is located in the outer zone of the medulla under normal physiologic conditions; their urine contains
and in the cortex. Cells lining the TAL are cuboidal with taurine and lacks tryptophan; and they also excrete creati-
eosinophilic cytoplasm and round central nucleus. Ultrastruc- nine in urine. Adult rabbits normally have cloudy urine
turally, cells of the TAL have prominent mitochondria, rough because most of their dietary calcium is absorbed and they
endoplasmic reticulum, and Tamm-Horsfall protein, which excrete it via the kidneys.
4 International Journal of Toxicology XX(X)

Table 1. Kidney Comparative Anatomy and Physiology.

Glomerular filtration Outer stripe of the


Species Nephron number (000s) rate (GFR; mL/min/kg) Medullary structure outer medulla Number of papillae

Human *300-1,100 1.78 Simple Very thin Multiple


Monkey *103-123 2.08 Simple Thin Single
Dog *340-490 6.13 Simple Almost absent Single
Rat *30 5.24 Complex Well developed Single
Mouse *10 14.0 Simple Well developed Single

Kidney Physiology low-molecular-weight proteins in the proximal tubules. These


are returned to the circulation by the peritubular and vasa recta
Kidney Glomerular Physiology capillaries. Sodium reabsorption by the proximal tubule is an
The primary functions of kidneys are (1) maintenance and active process that is driven by Naþ/Kþ ATPase at the baso-
regulation of body’s fluid and electrolytes, (2) maintenance lateral cell surface membranes. Reabsorption of NaCl accounts
of extracellular fluid volume, (3) endocrine such as elaboration for most of the energy expenditure in the kidney. Obligatory
of hormones, (4) regulation of blood pH and pressure, (5) (ie, water is obligated to follow Naþ) water reabsorption is a
excretion of the waste products of metabolism, and (6) meta- passive process. Approximately two-thirds of calcium reab-
bolic activities.7 Adequate blood flow and perfusion affects sorption is passive in exchange for sodium. Approximately
normal kidney function and about 10% of oxygen consumption two-thirds of phosphate is reabsorbed in the proximal tubules.
occurs in the kidneys. Measurement of kidney function is deter- In the dog, 85% of bicarbonate reabsorption takes place in the
mined by glomerular filtration rate (GFR) that varies across proximal tubules. Reabsorption and degradation of macromo-
species (Table 1) and pathologies in afferent and efferent arter- lecules in the ultrafiltrate takes place in the pars convoluta of
ioles (eg, drug-induced vasoconstriction) can affect GFR. The the proximal tubule due to its well-developed endocytic-
plasma is filtered via the glomerulus to produce glomerular lysosomal apparatus.2
ultrafiltrate. Ultrafiltration movement across the glomerulus Maintaining nearly constant blood plasma osmolality and
blood capillary network barrier into the urinary Bowman cap- sodium concentration is essential for survival. The urine con-
sule space involves crossing 3 layers: fenestrated endothelium centrating mechanism plays an essential role in regulating
of glomerular capillaries, GBM, and slit pores between the foot water and sodium excretion. The descending and ascending
processes of visceral epithelial cells of the Bowman capsule. portions of the loop of Henle enable recovery of much of the
The fenestrated (ie, have pores) endothelium allows the pas- Naþ and water that were filtered by the glomerulus. The urine
sage of fluid, blood plasma solutes, and low-molecular-weight concentrating and urine forming process in the kidney starts in
proteins and prevents passage of blood cells and platelets. Fac- the descending loop of Henle and depends on the generation of
tors that affect GBM glomerular filtration include molecular an interstitial osmotic gradient from the corticomedullary bor-
size, shape, and charge. Only small-molecular-weight mole- der down to the papilla tip. This osmotic gradient is generated
cules can pass through the glomerular filter and small mole- by a countercurrent (the descending and ascending loops fluid
cules (eg, amino acids, glucose, sodium, potassium, water) pass movement in opposite directions) multiplication (solute pump-
through the glomerular ultrafiltrate. Podocytes provide struc- ing leading to multiplication of solutes concentration in the
tural support to the capillary loop with their actin cytoskeleton, deep medulla) system within the descending and ascending
can express Toll-like receptor-4 (TLR4), and maintain GBM. loops of Henle, and passive diffusion in the vasa recta, the
There are structural proteins (eg, nephrin, podocin, and P cad- countercurrent exchanger. Cells in the descending thick loop
herin) involved in maintaining the structure of the foot pro- (DTL) have aquaporin (AQP) channel proteins (upper 40% of
cesses, and mutations in these proteins can lead to
each DTL expresses AQP1) to allow for the high permeability
proteinuria. Mesangial cells are phagocytic and may prevent
and movement of water from the descending loop lumen into
basement membrane accumulation of macromolecules that
the surrounding interstitium, leading to osmolality increases
escaped from the capillaries. The plasma ultrafiltrate passes
from approximately 300 mOsmol/kg (isosmotic with blood)
down the nephron tubule to form urine.2
in the cortex to about 1,200 mOsmol/kg (hypertonic solution)
in the papillary tip region, resulting in hypertonic
interstitium.3,8
Kidney Tubular Physiology There are important species differences in such urine osmol-
Physiologically, the proximal tubules account for 70% of all alities with toxicological implications and human risk assess-
reabsorptive activity. The glomerular ultrafiltrate undergoes ment if there are solubility issues with a test article. For
reabsorption process of sodium (65%), water (65%), potassium example, rats can concentrate to approximately 3,000 mOsm/kg
(65%), calcium (90%), phosphate, bicarbonate, magnesium, H2O and mice to approximately 4,300 mOsm/kg H2O. These
amino acids (99%), glucose (100%), vitamins, sulfate, and changes in urine osmolalities are accomplished by osmosis in
Radi 5

the descending limb and active epithelial cell transport in the factor (HIF) is a key regulator of adaptive responses in renal
ascending limb and lead to the reabsorption of up to 20% of the hypoxic conditions to regulate renal oxygen homeostasis, espe-
water and 10% of Naþ entering this segment of the nephron. cially in ischemia–reperfusion injury of acute kidney injury
Solutes and water recovered from these loops are returned to the (AKI).12 Renin is produced within juxtaglomerular (JG) cells
circulation via the vasa recta. The ascending loop of Henle lacks after processing and cleavage of circulating prorenin, which is
AQP proteins and is impermeable to water but is permeable to produced in the liver. Reduced renal perfusion pressure and
sodium and other solutes which are actively pumped out of the hyponatremia are the primary stimuli for renin release from
loop via the Naþ/Kþ ATPase pump. This leads to the hyper- JG cells that act as sensors of blood pressure. Renin is released
osmotic environment in the kidney medulla. The interstitium is by the sympathetic nervous system and AII, ADH, endothelin,
rich in osmotically active urea because of reabsorption by the and prostaglandins. Renin activates AII, which causes secretion
collecting ducts.8 of aldosterone by the adrenal cortical zona glomerulosa. The
The distal convoluted tubule plays a critical role in sodium, net effect is systemic vasoconstriction, intrarenal vasoconstric-
potassium, and cation homeostasis. It reabsorbs approximately tion, and increased aldosterone release. Thus, due to the
5% to 10% of the filtered sodium, 7% to 10% of filtered cal- reduced negative feedback on renin production by JG cells,
cium, 10% of filtered magnesium, and 10% to 15% of water. angiotensin-converting enzyme (ACE) inhibitors can cause
Water reabsorption in the distal tubules is under the influence hypertrophy and hyperplasia of JG cells. In addition, the kidney
of antidiuretic hormone (ADH; vasopressin). In addition, this is a site of degradation of hormones, such as insulin and aldos-
distal part of the nephron is aldosterone sensitive, a hormone terone, and is also responsible for the production of the active
that responds to volume depletion or hyperkalemia.9 Aldoster- form of vitamin D derived from vitamin D3 under the influence
one increases the amount of Naþ/Kþ ATPase in the basal mem- of parathyroid hormone.2 Kidney interstitial cells such as stel-
brane of distal tubules and Na/Cl reabsorption. Distal tubules late cells, in conjunction with pericytes, act as sensor and have
have receptors for parathyroid hormone to facilitate Caþþ endocrine effector functions for the control of blood pressure,
recovery. Hydrogen ions, creatinine, and some drugs such as salt balance, and oxygen delivery.13
penicillin are actively secreted and move from the blood into
the distal tubules.
The collecting ducts regulate urine volume and osmolarity
Kidney Metabolic Physiology
and contain 2 distinct cell types: principal cells and interca- The kidney is the primary route of xenobiotic excretion and
lated cells. Principal cells help maintain body water and salt contains xenobiotic biotransformation enzymes. This is
balance. Plasma osmolarity is regulated by the posterior pitui- because: (1) xenobiotics present in the glomerular ultrafiltrate
tary hormone ADH. Aldosterone is secreted by the adrenal are concentrated and present at high concentrations, (2) the
cortex in response to AII, a vasoconstrictor, stimulation. Prin- cortex receives about 80% of total renal blood flow and thus
cipal cells have receptors for ADH and aldosterone to regulate is exposed to the high concentration of xenobiotics coming
water and Naþ recovery. In fact, the principal cell plays a from the bloodstream, and (3) such high concentrations of
central role in salt and water transport with transporters such xenobiotics may accumulate in proximal tubulars following
as the epithelial Naþ channel (ENaC), the renal outer medul- reabsorption or secretion processes. Xenobiotic-metabolizing
lary Kþ channel, and the AQP2 water channel.10 Control of enzymes activities such as mixed-function oxidases are present
plasma Naþ and Kþ concentrations, extracellular fluid vol- in the proximal tubules and the activity varies along various
ume, and blood pressure is achieved via the coordinated reg- segments of the nephron.14 Mixed-function oxidase activity
ulation of ENaC by aldosterone and AQP2 by arginine occurs primarily in pars recta region of the proximal tubules
vasopressin in principal cells.10 Intercalated cells secrete or in the rat. The kidney plays an important role in the hydroxyla-
absorb acid or bicarbonate and play an important role in reg- tion and glucuronidation of fatty acids, arachidonic acid (AA),
ulating blood pH. Intercalated cells reabsorb Kþ and HCO3 and 25-hydroxyvitamin D3.15 Epoxidation of AA is catalyzed
and secrete Hþ. This function lowers the acidity of the plasma by the cytochrome P4502C (CYP2C) and CYP4F subfamilies
while increasing the acidity of the urine. and these enzymes play an important role in the renal regula-
tion of salt and water balance and in inflammation. The highest
activities of cytochromes P450 are present in the proximal
Kidney Endocrine Functions tubules and involved in conjugation of glutathione, glucuronic
The kidneys can be hormonally sensitive as they have high acid, and sulfate.16 The interspecies and sex differences in
levels of sex hormone receptor expression. Kidney endocrine kidney physiology among experimental laboratory animals in
function involves secreting 3 key hormones: erythropoietin, preclinical safety testing versus humans need to be considered
calcitriol (1,25-dihydroxycholecalciferol), and renin. In addi- when evaluating and interpreting test article–related kidney
tion, kidneys synthesize prostaglandins, growth factors, and changes and pathologies. S1, S2, and S3 tubular toxicological
arachidonate metabolites.2,6,11 Erythropoietin is produced by manifestation vary due to differences in kidney metabolic
renal cortical peritubular interstitial cells in response to activities, and there is little to no induction of cytochrome
hypoxic conditions. This stimulates erythropoiesis and the pro- P450s following administration of phenobarbital due to the
duction of red blood cells in bone marrow. Hypoxia-inducible absence of the CYP2B subfamily. In human kidney, CYP2D6
6 International Journal of Toxicology XX(X)

and CYP2E1 are absent. The activity of aldehyde oxidase in parenchyma and its various compartments via glomerular fil-
humans is higher than that in rats and dogs. Mitochondria in the tration or peritubular capillaries microcirculation.12,19,20 For
proximal tubules are the site of production of circulating 1a,25- example, lipopolysaccharides (LPS) are filtered into the tubular
dihydroxyvitamin D3.15 Because the blood flow in the renal fluid and can directly interact with renal tubular epithelial cells
medulla is much lower than the cortex, the levels of xenobiotic- (RTECs) via a TLR4-dependent mechanism.19,21 As a conse-
metabolizing enzymes are lower, and this region have more quence, various compartments of the kidney, including RTECs,
anaerobic metabolism. Some isoenzymes of P450 and glu- glomerular, and endothelial, are injured. This is because the
tathione S-transferases are localized in the thick ascending limb filtered PAMPs and DAMPs, also called “danger signals,” bind
and distal tubules, while prostaglandin H-synthase is localized to and activate RTECs, renal endothelial, and glomerular cells
in the collecting ducts in the medulla.17 Renal enzyme activi- and can lead to endothelial cell activation and structural
ties can be different between sexes and cytochrome P450 activ- changes in the glomerular and peritubular capillaries.20 There-
ity in rodents is often polymorphic by sex and sex hormones fore, exogenous and endogenous PAMPs and DAMPs are inte-
(eg, drug-induced acetaminophen toxicity) might influence gral to AKI pathogenesis.12
proximal tubules metabolizing enzyme activity. For example,
male mice have more P450 activity than female mice. Induc-
Clinical and Anatomic Pathology in Kidney
tion of kidney metabolizing enzymes activities can lead to
increases in kidney absolute weights.2
Safety Assessment
In addition, kidney transporters are important consideration Assessment for test article–related kidney pathology and toxi-
because interference with transporters by a drug candidate (eg, city can include standard clinical pathology parameters such as
inhibition of tubular secretion of creatinine without affecting clinical chemistries for blood urea nitrogen (BUN) and serum
glomerular function or causing any microscopic changes in the creatinine (sCr), along with disturbances in electrolytes and
kidney; drug is a substrate for a transporter; transporter affect- water balance, urine excretion of protein (eg, proteinuria, urine
ing drug disposition) might allow for mechanistic understand- protein to creatinine ratio, and microalbuminuria in nonrodents
ing of kidney safety signal risk assessment. For example, sex with glomerular pathologies) and electrolytes, urinalysis (urine
differences in transporter expression were observed with high- volume [eg, oliguria in AKI], appearance, specific gravity, pH,
est in the mouse followed by rat and dog. Organic anion trans- quantitation for microalbumin, and absolute and relative kid-
porter 2 was higher in male mouse than the female. Organic ney weight determination), and macroscopic and microscopic
cation transporter 2 was higher in male rat and mouse. Multi- examination of the kidney tissue.7,12 Microscopic examination
drug resistance protein 4 is predominantly expressed in male of urine sediment for presence of RBC, glucose, ketones, bilir-
mouse kidney.18 ubin, and crystals can be used to assess for drug-induced
changes such as crystalluria and it is important to consider
species differences in kidney drug handling. For example,
Kidney Pathophysiology and Toxicology drug-induced crystalluria can be noted in rats without histo-
pathological evidence of kidney injury. In addition, urinary
Kidney Pathophysiologic Responses bladder tumors if noted in rodents due to crystalluria mechan-
The kidney is a common target organ of toxicity and injury as a ism of action (MOA) are not relevant to human cancer risk, and
result of its exposure to drugs, xenobiotics, or chemicals. This rodent urothelial proliferative lesions secondary to crystalluria
is because: (1) the high blood flow rate as they receive more are not predictive of human cancer risk. Furthermore, rats are
blood flow per gram of tissue (approximately 25% of cardiac sensitive to the development of irritation-related urothelial
output) than any other organ, (2) high doses of test articles tumors as a result of crystalluria, while mice are much less
typically administered in preclinical toxicity and safety studies, likely to develop urothelial tumors secondary to crystals or
(3) kidney’s key role in excretion through urine, (4) high renal uroliths. Drug-induced kidney glomerular pathological
metabolic biotransformation and high oxygen consumption of changes can manifest clinically as nephrotic syndrome due to
renal epithelium, (5) high transporter activity of the renal albumin loss (ie, plasma proteins leaks into the urine [protei-
epithelium, and (6) kidney’s role in reabsorption and its ability nuria] through abnormally permeable GBM). Such reduction in
to concentrate fluids.2,7 The specific pattern of kidney toxicity plasma osmotic pressure leads to edema, pleural effusions,
and injury is dependent on the physiochemical properties of the reduced intravascular volume, renal hypoperfusion, and/or sec-
drugs (eg, low intrinsic solubility) and their dose, xenobiotics, ondary hyperaldosteronism. Drug-induced kidney glomerular
or chemicals, toxicokinetic properties, renal clearance profile pathological changes can also lead to nephritic syndrome,
and metabolic attributes, and local kidney tissue concentration which usually presents with hematuria, red cell casts in the
and length of time of exposure. In addition, the kidney par- urine, azotemia, and/or oliguria. Toxicologists need to be
enchyma is especially vulnerable to the pro-inflammatory familiar with this language to appropriately communicate pre-
response to infection and subsequent kidney tissue damage. clinical kidney findings with nephrologists and clinicians.
This is because damage-associated molecular patterns For urine biomarkers, it is essential to collect urine speci-
(DAMPs), pathogen-associated molecular patterns (PAMPs), mens over a 16- to 24-hour period, ideally in metabolism cages
and pro-inflammatory mediators gain access to kidney on cold packs. For investigative toxicological assessment of
Radi 7

Table 2. Kidney Spontaneous/Incidental Pathologies in Experimental Laboratory Animals.

Species Glomerular Tubular Interstitial Vascular

Monkey Fatty metaplasia, immature glomerulus, Mineralization Cell infiltrates Arteritis, polyarteritis
sclerosis nodosa
Minipig Glomerulonephritis, hemorrhage, and Hyaline casts, acute tubular hemorrhage, Cell infiltrates Arteritis and vasculitis
sclerosis hyaline droplets
Dog Focal/segmental lipidosis/foam cells, focal Mineralization, hyaline casts, vacuolation Cell infiltrates Idiopathic necrotizing
glomerulitis, immature glomerulus polyarteritis
Rat Focal/segmental glomerular sclerosis Mineralization, hyaline casts, dilatation, Lipomatous Mineralization,
hydronephrosis and cysts (juvenile rats) metaplasia polyarteritis nodosa
Mouse Amyloidosis Hyaline casts, dilatation, vacuolation Lipomatous Polyarteritis
metaplasia

rodent crystalluria, it is important not to fast the animals before glomeruli in the cortex of dogs2 and monkeys, corticomedul-
urine collection. Historical control data from the testing facility lary mineralization or nephrocalcinosis in female rats, kidney
might be used to help distinguish test article-related effects pelvis dilatation in rats, kidney pelvis minimal epithelial hyper-
from background changes and in assessing individual animal plasia and mineralization in rats, increased hyaline droplet
variability. This is especially important in rats and mice accumulation in rats, tubular epithelial vacuolation in mice,
because renal function tests may become more variable due mineral deposits, and interstitial mononuclear cell infiltrates
to development of spontaneous age-related, species-specific in multiple species (Table 2). Mineral deposits, for example,
kidney pathologies with no human counterpart such as chronic occur at the corticomedullary junction in female rats due to
progressive nephropathy (CPN) as early as 12 weeks of age.2 shedding of microvilli and microvesicles from S1 proximal
Rats with CPN may show increases in kidney weights; tubules22 and accumulation in the outer stripe of medulla.7 In
increased urine excretion of protein and calcium; decreased beagle dogs, such deposits can be noted in papillary collecting
urinary excretion of sodium and chloride; decreased creatinine duct region and in the interstitium of renal papilla in cynomol-
clearance; increased serum concentration of BUN, sCr, gus monkeys. The CPN is a spontaneous disease of laboratory
sodium, and chloride; and decreased serum concentration of rats and mice that represents a significant confounder for inter-
albumin. Castration can result in a lower incidence and severity pretation of kidney histopathology in toxicology studies after
of CPN. The physiologic status of experimental laboratory ani- test article administration. Male gender is a primary risk factor
mals, diet (eg, imbalance of calcium/phosphorus leading to for developing CPN. The CPN is an age-related disease in rats
mineralization in rats), and various physiologic parameters and mice influenced by numerous extrinsic factors such as diet
such as age, sex, and interspecies differences in kidney anat- and chemical exposures.2 Dietary restriction prevented or
omy and physiology are important when assessing kidney delayed the development of glomerulosclerosis, tubulointersti-
responses to injurious test articles and/or chemicals.2,7
tial damage, functional changes, morbidity, and mortality asso-
ciated with CPN in rats by controlling initial body and kidney
Kidney Spontaneous Pathologies growth, glomerular size, and nephron hypertrophy.2,23 Micro-
Histopathology is recognized as the single most appropriate scopically, there is tubular atrophy, interstitial fibrosis, tubular
screen for evidence of kidney injury. Familiarity with sponta- dilation, casts, hyperplasia, dilated Bowman space, glomerulo-
neous diseases occurring in laboratory animals is essential for sclerosis, glomerular atrophy, casts, and interstitial inflamma-
the establishment of the nature and cause of morbidity or mor- tory infiltrates.2,7
tality in an individual animal. For example, rats, especially Mature male rats spontaneously develop prominent hyaline
Sprague Dawley strain, have vault-like folds called fornices droplets in the cytoplasm of the S2 segment of proximal
in the upper portion of kidney pelvis that extend into the renal tubules. Ultrastructurally, these hyaline bodies consist of
medulla and as a result can develop age-related and species- alpha2m globulin and represent glomerular filtration and
specific spontaneous, random (ie, no dose–response relation- resorption and catabolism of low-molecular-weight proteins
ship), mineralization, inflammation, and/or hyperplasia in (eg, albumin and immunoglobulins) in secondary lysosomes.
males and/or females in this unique anatomic location. Alpha2m-globulin is androgen regulated and synthesized in the
Mechanisms for the development of urothelial proliferative liver of male rats only, which is rapidly cleared in the glomer-
lesions in rodents (ie, genotoxicity, mitogenicity, cytotoxicity) ular filtrate. Alpha2m-globulin is lost in the urine in the normal
need to be ruled out in the risk assessment paradigm. Most mature male rat, creating a physiologic proteinuria. Thus,
common spontaneous/incidental light microscopic changes in alpha2m-globulin nephropathy is a sex- and species-specific
control laboratory animals include CPN in rodents, glomerular entity that can be exacerbated by test article administration and
amyloidosis in CD-1 mice, glomerular lipidosis/foam cells in is not relevant to humans.2,7 Xenobiotics or their metabolites
beagle dogs with no effects on glomerular function, immature can reversibly bind to alpha2m-globulin and decrease
8 International Journal of Toxicology XX(X)

effectiveness of lysosomal degradation leading to accumula- negatively charged GBM, and podocytes. These features and
tion. The hyaline droplets can be readily visualized with histo- the presence of complement regulatory proteins are contribut-
chemical stains including the Mallory-Heidenhain stain. In ing factors in ICD. For example, the presence of in situ granular
addition, a wear-and-tear lipofuscin pigment can be noted in deposits of immunoglobulins and/or complement at the site of
the RTECs in rats and increases with age without kidney func- tissue injury in kidney sections, using immunohistochemical
tional sequela or clinical significance. Idiopathic necrotizing staining, even in few animals, is generally consistent with ICD.
polyarteritis (beagle pain syndrome) is a spontaneous condition In the kidney of humans and/or rodents, cell surface and soluble
that can be seen in beagle dogs. Clinically, it can be associated complement regulatory proteins (eg, decay accelerating factor
with fever, weight loss, and cervical pain (“beagle pain [CD55], membrane cofactor protein [CD47], complement
syndrome”). Pathologically, there is involvement of medium- receptor type 1 (CR1), CD59, clusterin, and Factor H) are
sized muscular arteries including those in the kidney leading to expressed by various microanatomic kidney compartments,
thickened, tortuous vasculature with fibrinoid necrosis, mixed including glomerular capillaries, peritubular capillaries, prox-
cell infiltrates, and thrombosis. Beagle pain syndrome is pre- imal tubules, collecting ducts, medullary interstitium, and glo-
sumed to be an immune-mediated condition. Other presenta- merular cells (endothelial, epithelial, and mesangial).12,26-28
tions of vasculitis in dogs might include dermatomyositis and Thus, immune complex-mediated type 3 hypersensitivity reac-
similar disorders with ischemic cutaneous and vascular lesions tions and ICD are relatively common in toxicology studies,
(ie, dermatopathy). especially with biotherapeutics after repeated dose administra-
tion to monkeys.29 The reader is strongly encouraged to refer to
comprehensive reviews on ICD pathophysiology, especially as
Drug-Induced AKI and Kidney Toxicology it relates collective weight-of-evidence approach, time course
of complement formation, and antidrug antibody (ADA) for-
Acute Kidney Injury mation and its correlation with exposure in toxicity studies.
Acute kidney injury is one of the most common causes of Immune complex-mediated glomerular pathology related to
kidney toxicity. In AKI, impairment in kidney function (eg, immunogenicity in animals is generally not considered relevant
rise in sCr and/or sustained oliguria) leads to retention of nitro- for predicting immunogenicity in humans. For example,
genous waste products. Pathophysiologically, the classification administration of obinutuzumab, anti-CD20 monoclonal anti-
of AKI can be divided into 3 categories: (1) prerenal, (2) intrin- body, in a 26-week toxicity study in cynomolgus monkeys
sic [renal], and (3) postrenal.12 In prerenal AKI, kidney perfu- resulted in dose-independent ICD-mediated hypersensitivity
sion decreases as a result of hypovolemia (eg, hemorrhage, and chronic inflammation in tissues including the kidneys.
vomiting, or diarrhea), decreased cardiac output (eg, heart fail- Kidney glomerulus pathological changes included mesangial
ure or pulmonary embolism), impairment of kidney vasomo- hypercellularity and thickening, proliferation of parietal
dulation (eg, nonsteroidal anti-inflammatory drugs [NSAIDs], epithelial cells in Bowman space with crescents formation,
ACE inhibitors, or cyclosporine), and/or vasodilation (eg, sep- and periglomerular mononuclear infiltrates. Subepithelial
sis or hepatorenal syndrome).12 Intrinsic AKI etiopathogenesis electron-dense deposits in glomerular capillary basement
could be related to specific insults in kidney vascular, glomer- membrane consistent with ICD were noted by transmission
ular, tubular, or interstitial compartments. Cellular insults in electron microscopy.30
intrinsic AKI can lead to various cell death modalities such In dogs, glomerulopathy and/or proteinuria has been
as apoptosis, autophagy, or regulated and genetically con- observed in dogs with Cushing syndrome, a syndrome caused
trolled cell death (necroptosis).24 Vascular causes of AKI by excessive adrenocorticotropic hormone production (hyper-
include vascular stenosis or vasculitis (eg, disseminated intra- adrenocorticism) and in dogs treated with prednisone. 25
vascular coagulation or drug induced). Glomerular AKI Increased urine volume, decreased urine specific gravity, pro-
includes immune complex diseases (ICD), glomerulonephritis, teinuria, and glomerulopathy were observed in 10 male beagle
and postinfectious glomerulonephritis. Tubular and interstitial dogs after oral administration of prednisone at 2.2 mg/kg,
AKI can be caused by ischemia (shock), inflammation (eg, twice daily, for 6 weeks. Similarly, drug-induced glomerulo-
sepsis, infections), rhabdomyolysis, hemolysis, crystals, ethy- pathy was observed microscopically in dogs in a 9-month
lene glycol (EG), cisplatin toxicity, or medications (eg, amino- toxicity study after oral administration of fosdagrocorat, a
glycosides, rifampin, NSAIDs, prednisone).12,25 Postrenal AKI dissociated agonist of the glucocorticoid receptor, to beagle
can be caused by bladder obstruction or urinary stones. dogs. It is interesting to note that obesity resulting from ad
libitum feeding exacerbated the severity of the kidney find-
ings observed in both the fosdagrocorat- and prednisone-
Drug-Induced Kidney Glomerular Pathologies treated dogs, with the prednisone-treated dogs showing
As the initial plasma filtering portion of the nephron, the glo- greater overall glucocorticoid-induced renal effects than those
merulus plays an integral role in AKI and drug-induced injury dosed with fosdagrocorat.25
such as ICD. The glomerulus selective barrier to filtered mole- It is important to note that drug-induced minimal change
cules based on their size, shape, and/or charge is facilitated by disease (MCD) can be noted in various experimental laboratory
its structural anatomical features of fenestrated endothelium, a animals. The MCD is by characterized by normal glomerular
Radi 9

Table 3. Examples of Drugs That Induce Kidney Pathologies in Experimental Laboratory Animals.

Species Glomerular Tubular Interstitial Vascular

Monkey Human biotherapeutics, VEGFR2 NSAIDs, antisense oligonucleotide, NSAIDs Antisense deoxyoligonucleotide,
inhibitor, antisense recombinant IL-18 human biotherapeutics, oral
oligonucleotide, angiotensin II phosphodiesterase inhibitors
receptor antagonists
Minipig Cyclosporine Single-stranded oligonucleotide Cyclosporine Betamethasone, hydrocortisone
Dog Hydrocortisone, ampicillin, ACE inhibitors, tenofovir Cisplatin, estrogen Glucocorticoid agonists
growth hormone diphosphonate,
NSAIDs
Rat 5-lipooxygenase, human basic ACE inhibitors, aminoglycoside Cisplatin, NSAIDs Cyclosporine, oral
fibroblast growth factor, H2 antibiotics, cyclosporine, glycoprotein phosphodiesterase inhibitors,
receptor antagonists IIb-IIIa antagonists, fibrans, siRNA angiotensin II antagonist
oligonucleotide therapeutics
Mouse Antisense oligonucleotide Cisplatin, methotrexate, antisense ACE inhibitor ACE inhibitor, ephedrine
oligonucleotide
Abbreviations: ACE, angiotensin-converting enzyme; H2 receptor antagonists, histamine 2 receptor antagonists; IL, interleukin; NSAIDs, nonsteroidal
anti-inflammatory drugs; siRNA, small interfering RNA; VEGF, vascular endothelial growth factor.

morphology by light microscopic examination and lack of fibroblast growth factor to rats and monkeys. Histopathologically,
tubulointerstitial or vascular pathologies. However, animals enlargement, vacuolation, and karyomegaly of podocytes in glo-
can be proteinuric with nephrotic syndrome and glomerular meruli, hypertrophy/hyperplasia of the parietal epithelium of
changes of podocyte foot process effacement and protein Bowman capsule in the glomeruli, tubular dilatation and cast
resorption droplets, and cell lysis can be noted only by electron formation, thickening of the media in the lobular arteries, arterio-
microscopic examination. 5-Lipooxygenase inhibitors in rats pathy, and/or hyperplasia of the epithelium of the papilla and
and masitinib mesylate (tyrosine-kinase inhibitor) in dogs are collecting ducts were noted.34
examples of drug-induced MCD31,32 (Table 3). Toxicologists Anti-vascular endothelial growth factor (VEGF) therapies
need to consider alternative assessment methodologies such as can induce glomerular thrombotic microangiopathy, mild pro-
electron microscopy if kidney microscopic examination is teinuria, and hypertension in humans.35 Strong VEGF expres-
within normal limits, yet there is evidence of drug-induced sion in mice is demonstrable using immunohistochemistry
proteinuria. in glomerular podocytes.36 In addition, VEGF plays a role in
Drug-induced segmental glomerulosclerosis is rare in dogs glomerular endothelial cell fenestration formation, at least in
but has been noted in preclinical safety studies with adminis- vitro.37 The VEGF is released by podocytes and binds to its
tration of growth hormone and synthetic progesterone.33 Focal receptors on endothelial cells (eg, VEGFR-2). This helps main-
segmental glomerulosclerosis (FSGS) is a kidney disease in tain podocyte health and slit diaphragm integrity because inhi-
humans characterized by progressive glomerular scarring and bition of VEGF signaling downregulates the expression of
a clinical presentation of nephrotic syndrome. In primary nephrin, a protein involved in the maintenance of the glomer-
FSGS, immunologic podocyte injury takes place. In secondary ular slit diaphragm. Reduction in VEGF after administration of
FSGS, pathologic glomerular hyperfiltration/hypertension some anti-VEGF drugs (eg, VEGFR-2 inhibitor) can lead to
dose-dependent, yet reversible, glomerular filtration barrier
takes place in patients with an underlying etiology (eg, obesity,
injury in rats and/or monkeys after test article administration
diabetes mellitus). Examples of drug-induced FSGS-like
(Table 3). Histopathologically, eosinophilic, hyaline droplets
pathologies include Adriamycin, puromycin, and streptozoto-
can be noted in podocytes, mesangial, and/or endothelial cells
cin. In the early stages of FSGS-like pathologies, glomerular
and podocyte and parietal epithelial cell hypertrophy and
hypertrophy/enlargement and/or mesangial hypercellularity
mesangial cell hyperplasia. Similar droplets might be seen in
might be noted morphologically. Interestingly, glomerular tufts
the S3 segment of proximal tubules.
collapse and extracapillary proliferation with reticular inclu-
sions can be noted by electron microscopy with some viral
infections (eg, Simian immunodeficiency virus) or some drugs
[anabolic steroids and interferons]). Differentiating primary
Kidney Drug-Induced Tubular, Interstitial, and
versus secondary FSGS-like glomerular pathologies in experi- Vascular Pathologies
mental laboratory animals administered a test article is impor- The immunopathogenesis of AKI is an important consideration
tant in understanding drug MOA and differentiating primary as the kidneys contribute to the immune system homeostasis by
versus secondary glomerular insults. removing circulating cytokines and bacterial toxins (eg, LPS)
Drug-induced glomerular vacuolation and proteinuria has and by continuously sampling blood-borne proteins.38 Emer-
been noted after intravenous administration of human basic ging evidence supports the involvement of RTECs and the
10 International Journal of Toxicology XX(X)

innate and adaptive arms of the immune system in the patho- Examples of drug-induced tubular injury include aminogly-
genesis of intrinsic AKI. Pro-inflammatory DAMPs, PAMPs, coside antibiotics (eg, gentamicin), cephalosporins, cisplatin,
TLRs, oxidative stress, HIF, complement system, adhesion antiviral agents, and calcineurin (CNI) inhibitors (Table 3).
molecules, cell death, resident renal DCs, neutrophils, T and Pathophysiologic factors in aminoglycoside kidney toxicology
B lymphocytes, macrophages, natural killer T cells, secreted include alterations in tubular cells reabsorption, tubular
cytokines, and chemokines contribute to the immunopathogen- obstruction and tubular malfunction, reduced glomerular filtra-
esis of AKI. Thus, loss of the immune system homeostasis can tion, tubuloglomerular feedback activation, renal vasoconstric-
have direct or indirect effects on kidney pathology. If the tion, and mesangial contraction.42 Local kidney factors (eg,
immunopathologic processes in AKI continue, this can lead variability in P-glycoprotein and CYP3A4/5 expression or
to renal fibrosis and/or chronic kidney disease.12 activity, older age, salt depletion, use of NSAIDs, and genetic
Sepsis is one of the most common causes of AKI.39 In polymorphisms in genes) are important for susceptibility CNI
sepsis, there is a systemic inflammatory response, triggered kidney toxicity.43
by the innate immune system, to attack foreign bacterial patho- Unless associated with morphologic evidence of tubular cell
gens (eg, LPS) and toxins to limit their spread.20 Such systemic injury of necrosis and/or degeneration, antisense oligonucleo-
inflammatory response leads to the release of PAMPs and tides (ASO) can cause nonadverse and adaptive kidney changes
DAMPs into the circulation.20 The PAMPs are released from in rats evident histologically by accumulation of basophilic or
pathogens and DAMPs are released from damaged and injured eosinophilic and granular, amorphous, or vacuolar material in
tissues at sites of infection, and both PAMPs and DAMPs the cytoplasm or nuclei of renal epithelial cells. This is con-
prime and signal the immune system to fight back the invading sistent with lysosomal accumulation and endosomal localiza-
pathogens and toxins. Interactions of PAMPs and DAMPs with tion of ASO. Oligonucleotides can be absorbed rapidly and
sensors of the innate immune system called pattern recognition distributed in tissues, most commonly in the kidneys, after
receptors (eg, TLRs, nucleotide-binding oligomerization systemic or local administration. In rats, exacerbation of CPN
domain-like receptors, and retinoic-acid-inducible-gene-I and proteinuria might be observed after ASO chronic adminis-
[RIGI]-like receptors) amplify the systemic inflammatory tration. Pathophysiologically, once oligonucleotides interact
response.12,19,20 For example, there is TLR4-dependent LPS with and taken by the brush boarder of proximal tubules via
recognition mechanism in the RTECs in the S1 segment of the polyanion transporter, they are internalized by endocytosis,
proximal tubules.21 Kidney-specific DAMPs (eg, uromodulin leading to incorporation into endosomes and eventually lyso-
[Tamm-Horsfall protein]) are synthesized by distal epithelial somes and phagolysosomes. Some reports suggest that
tubules and selectively released into the tubular lumen and leak although there are high concentrations of ASO and cytoplasmic
into the interstitium after renal tubular injury. This activates vacuolation, no evidence of kidney functional abnormalities
renal DCs via TLR4, and NACHT, LRR, and PYD domains- (eg, sCr or BUN) were noted, but low incidence of reversible
containing protein 3 inflammasome potentially leading to ster- increase in urine protein to creatinine ratio was also noted in
ile kidney inflammation.40,41 monkeys.44
The kidney parenchyma is especially vulnerable to the pro- Drugs that can induce crystal nephropathy in humans and/or
inflammatory response to infection and subsequent kidney experimental animals include quinolone antibiotics (eg, ampi-
tissue damage because it receives approximately 25% of the cillin, norfloxacin, and ciprofloxacin), sulfonamides and fluor-
cardiac output. This is because the toxin-rich blood, PAMPs, oquinolones, glycoprotein IIb-IIIa antagonists, ganciclovir,
DAMPs, and pro-inflammatory mediators gain access to kid- sulfadiazine, and dopamine autoreceptor agonist (Table 3).
ney parenchyma and its various compartments via glomerular Intratubular precipitation of xenobiotics/crystals can cause or
filtration or peritubular capillaries microcirculation.19,20 For promote AKI and chronic kidney injury. A number of xenobio-
example, LPS is filtered into the tubular fluid and can directly tics/drugs are known to precipitate within renal tubules, due to
interact with RTECs via a TLR4-dependent mechanism.19,21 their insolubility (parent drug or its metabolite) within urine.
As a consequence, various compartments of the kidney includ- Risk factors for xenobiotic/crystal precipitation within the kid-
ing RTEC, glomerular, and endothelial are injured. This is ney tubules include intravascular volume depletion, underlying
because the filtered PAMPs and DAMPs, also called “danger kidney disease, and metabolic disturbances that promote
signal,” bind to and activate RTECs and kidney endothelial and changes in urine pH. EG-mediated crystal nephropathy
glomerular cells and can lead to endothelial cell activation and requires metabolic transformation of EG to oxalic acid that
structural changes in the glomerular and peritubular capil- takes place primarily in the liver via alcohol dehydrogenase.
laries.20 It is important to distinguish adaptive kidney response When oxalic acid is concentrated in the urine, calcium oxalate
(eg, drug-induced medullary tubular hypertrophy, tubular crystals form, leading to crystalluria, nephrolithiasis, metabolic
hypertrophy secondary to increased GFR, potassium depletion acidosis, low urine specific gravity, and proteinuria. The EG-
[chronic hypokalemia], high dietary sodium chloride, increased induced crystal deposition in kidney proximal tubules was
sodium loss, testosterone treatment, adrenocorticotropic detected unbound in the lumen and attached to the surface of
hormone-mediated hyperadrenocorticism1,2) from direct injur- epithelial cells.45 Crystal formation can lead to chronic inter-
ious tubular damage. Collectively, exogenous and endogenous stitial nephritis with glomerulosclerosis, tubular atrophy, and
PAMPs and DAMPs are integral to AKI pathogenesis. interstitial fibrosis. 46 Sulfonamides have caused crystal
Radi 11

nephropathy, affecting the distal nephron in animals and employed on a case-by-case basis to further dissect a drug-
humans. As the pH of murine urine generally varies between induced kidney finding.
5 and 8, and is greatly influenced by diet, this would influence
crystal nephropathy formation. In addition, a major difference Conclusions
between murine and human urine is the overall osmolality.
Murine urine is usually highly concentrated, with male urine Detection and characterization of direct and indirect kidney
having higher protein concentrations than females. It is impor- changes and/or toxicity in preclinical species and human rele-
tant to note that for human risk assessment, murine urinary vance will depend on the preclinical safety testing strategy and
crystalluria MOA is not relevant to human urinary bladder collective weight-of-evidence approach including new investi-
gational drug MOA, drug physiochemical properties, distin-
cancer risk.
guishing effects related to the parent drug or its metabolite,
Cyclooxygenase-mediated prostaglandins are involved in
changes in sCr related to renal transporters, preclinical and
renal functions and their inhibition is associated with potential
clinical interspecies, and sex differences in kidney macro- and
adverse renal effects.11 Analgesic nephropathy is characterized
micro-anatomy, physiology, and metabolic and hormonal
by chronic interstitial nephritis and renal papillary necrosis
activities, background pathologies, drug-induced, yet sex-
(RPN) or calcifications. Some laboratory animals (eg, rats,
and/or species-specific kidney toxicity, and MOA relevance
dogs) are unusually susceptible to RPN. Kidney changes in
to humans. Various investigative toxicology and pathology
monkeys associated with administration of NSAIDs-
tools can be employed on a case-by-case scientific basis to
containing carboxylic acid, such as naproxen, and carboxylic
better characterize pharmaceutical drug-induced kidney
acid moiety (eg, fibrans) were shown to induce formation of
toxicities.
casts with crystalline material that precipitates in the kidneys.29
This can lead to obstructive nephropathy in rats because of Author Contributions
accumulation of intratubular crystal precipitates (eg, glycopro-
Z. Radi contributed to conception and design, contributed to acquisi-
tein IIb-IIIa antagonists).46 Obstructive nephropathy can be
tion, analysis, and interpretation, drafted the manuscript, critically
seen secondary to co-precipitation of administered IL-18 and revised the manuscript, gave final approval, and agrees to be accoun-
Tamm-Horsfall protein in the distal nephron (Table 3).47 table for all aspects of work ensuring integrity and accuracy.
Drug-induced inclusion bodies can be noted in rats with
norepinephrine/serotonin reuptake inhibitors and propiverine Declaration of Conflicting Interests
(anticholinergic drug). Such inclusion bodies can be related The author(s) declared the following potential conflicts of interest
to D-amino acid oxidases (DAO). The physiologic function of with respect to the research, authorship, and/or publication of this
DAO, present in the kidney, liver, and brain, is to act as a article: The author is a Pfizer Inc. employee.
detoxifying agent and to metabolize D-amino acids. The patho-
physiologic mechanism of DAO nuclear and cytosolic accumu- Funding
lation in kidney tubules in the form of inclusion bodies is likely The author received no financial support for the research, authorship,
related to peroxisome proliferation and enzyme induction and/or publication of this article.
mediated through peroxisome proliferator activated receptor-
a (PPARa).48 The PPARa is found predominately in the renal References
cortex proximal convoluted tubules49 and increased DAO 1. Haschek WM, Rousseaux CG, Wallig MA. Fundamentals of
activity has been demonstrated in kidneys of rats exposed to Toxicologic Pathology. Houston, TX: Gulf Professional Publishing;
ciprofibrate, a PPARa agonist.50 The kidney finding of tubular 2010.
inclusion bodies is species-specific, reversible, and not relevant 2. Khan N, Hard GC, Alden CL. Kidney. Haschek and Rousseaux’s
to human safety risk assessment.48 Other examples of drug- Handbook of Toxicologic Pathology. California, United States:
induced kidney (vascular and interstitial) injury are outlined Academic Press; 2013.
in Table 3. Also, there are species differences in response to 3. Fogo AB, Cohen AH, Colvin RB, Jennette JC, Alpers CE. Funda-
drug-induced injury. For example, while rats and monkeys are mentals of Renal Pathology. Berlin, Germany: Springer; 2014.
sensitive to the development of kidney pathology after puro- 4. Kriz W. Structural organization of the renal medulla: comparative
mycin administration, dogs, mice, and rabbits are relatively and functional aspects. Am J Physiol. 1981;241(1):R3-R16.
insensitive. Finally, various investigative techniques (eg, 5. Tsuchiya N, Matsushima S, Takasu N, Kyokawa Y, Torii M.
immunohistochemistry, histochemical stains; in situ hybridi- Glomerular calcification induced by bolus injection with dibasic
zation, polarized light microscopic examination to visualize sodium phosphate solution in Sprague-Dawley rats. Toxicol
crystals in kidney sections, monocyte chemoattractant Pathol. 2004;2(4):408-412.
protein-1 and C-reactive protein biomarkers for vascular 6. Zeisberg M, Kalluri R. Physiology of the renal interstitium. Clin J
pathologies, complement levels, inulin clearance, endogenous Am Soc Nephrol. 2015;10(10):1831-1840.
creatinine clearance, kidney microautoradiography, confocal 7. Khan N, Hard G, Radi Z. Renal toxicity. In:Wilson AGE, ed.
Raman microspectroscopy, matrix-assisted laser desorption/ New Horizons in Predictive Toxicology: Current Status and
ionization mass spectrometry imaging).2,7 might need to be Application; 2012: 499-541.
12 International Journal of Toxicology XX(X)

8. Dantzler WH, Layton AT, Layton HE, Pannabecker TL. Urine- 26. Ichida S, Yuzawa Y, Okada H, Yoshioka K, Matsuo S. Localiza-
concentrating mechanism in the inner medulla: function of the tion of the complement regulatory proteins in the normal human
thin limbs of the loops of Henle. Clin J Am Soc Nephrol. 2014; kidney. Kidney Int. 1994;46(1):89-96.
9(10):1781-1789. 27. Nangaku M. Complement regulatory proteins in glomerular dis-
9. Subramanya AR, Ellison DH. Distal convoluted tubule. Clin J Am eases. Kidney Int. 1998;54(5):1419-1428.
Soc Nephrol. 1014;9(12):2147-2163. 28. Thurman JM, Ljubanovic D, Royer PA, et al. Altered renal tub-
10. Pearce D, Soundararajan R, Trimpert C, Kashlan OB, Deen PM, ular expression of the complement inhibitor Crry permits comple-
Kohan DE. Collecting duct principal cell transport processes and ment activation after ischemia/reperfusion. J Clin Invest. 2006;
their regulation. Clin J Am Soc Nephrol. 2014;10(1):135-146. 116(2):357-368.
11. Radi ZA. Pathophysiology of cyclooxygenase inhibition in ani- 29. Leach MW, Frank DW, Berardi MR, et al. Renal changes asso-
mal models. Toxicol Pathol. 2009;37(1):34-46. ciated with naproxen sodium administration in cynomolgus mon-
12. Radi ZA. Immunopathogenesis of acute kidney injury. Toxicol keys. Toxicol Pathol. 1999;27(3):295-306.
Pathol. 2018; 46(8):930-943. 30. Husar E, Solonets M, Kuhlmann O, et al. Hypersensitivity reac-
13. Kurtz A. Endocrine functions of the renal interstitium. Pflügers tions to obinutuzumab in cynomolgus monkeys and relevance to
Arch. 2017;469(7-8):869-876. humans. Toxicol Pathol. 2017;45(5):676-686.
14. Nigam SK, Wu W, Bush KT, Hoenig MP, Blantz RC, Bhatnagar 31. Read NG, Astbury PJ, Evans GO, Goodwin DA, Rowlands A.
V. Handling of drugs, metabolites, and uremic toxins by kidney Nephrotic syndrome associated with N-hydroxyureas, inhibitors
proximal tubule drug transporters. Clin J Am Soc Nephrol. 2015; of 5-lipoxygenase. Arch Toxicol. 1995;69(7):480-490.
10(11):2039-2049. 32. Brown MR, Cianciolo RE, Nabity MB, Brown CA, Clubb FJ,
15. Wang Y, Zhu J, DeLuca HF. The vitamin D receptor in the Lees GE. Masitinib-associated minimal change disease with acute
proximal renal tubule is a key regulator of serum 1a,25- tubular necrosis resulting in acute kidney injury in a dog. J Vet
dihydroxyvitamin D3. Am J Physiol Endocrinol Metab. 2015; Inter Med. 2013;27(6):1622-1626.
33. Prahalada S, Stabinski LG, Chen HY, et al. Pharmacological and
308(3):E201-E205.
toxicological effects of chronic porcine growth hormone admin-
16. Lock EA, Reed CJ. Xenobiotic metabolizing enzymes of the kid-
istration in dogs. Toxicol Pathol. 1998;26(2):185-200.
ney. Toxicol Pathol. 1998;26(1):18-25.
34. Mazué G, Newman AJ, Scampini G, et al. The histopathology of
17. Smith WL, Wilkin GP. Immunochemistry of prostaglandin
kidney changes in rats and monkeys following intravenous admin-
endoperoxide-forming cyclooxygenases: the detection of the
istration of massive doses of FCE 26184, human basic fibroblast
cyclooxygenases in rat, rabbit, and guinea pig kidneys by immu-
growth factor. Toxicol Pathol. 1993;21(5):490-501.
nofluorescence. Prostaglandins. 1977;13(5):873-892.
35. Pfister F, Amann K, Daniel C, Klewer M, Büttner A, Büttner-
18. Basit A, Radi Z, Vaidya V, Karasu M, Prasad B. Kidney cortical
Herold M. Characteristic morphological changes in anti-VEGF
transporter expression across species using quantitative proteo-
therapy-induced glomerular microangiopathy. Histopathology.
mics. Drug Metab Dispos. 2019. Manuscript submitted.
2018;73(6):990-1001.
19. Gomez H, Ince C, De Backer D, et al. A unified theory of sepsis-
36. Maharaj AS, Saint-Geniez M, Maldonado AE, D’Amore PA.
induced acute kidney injury: inflammation, microcirculatory dys-
Vascular endothelial growth factor localization in the adult. Am
function, bioenergetics, and the tubular cell adaptation to injury.
J Pathol. 2006;168(2):639-648.
Shock. 2014;41(1):3-11. 37. Esser S, Wolburg K, Wolburg H, Breier G, Kurzchalia T, Risau
20. Mårtensson J, Bellomo R. Pathophysiology of septic acute kidney W. Vascular endothelial growth factor induces endothelial fenes-
injury. Contrib Nephrol. 2016;187:36-46. trations in vitro. J Cell Biol. 1998;140(4):947-959.
21. Kalakeche R, Hato T, Rhodes G, et al. Endotoxin uptake by S1 38. Tecklenborg J, Clayton D, Siebert S, Coley SM. The role of the
proximal tubular segment causes oxidative stress in the down- immune system in kidney disease. Clin Exp Immunol. 2018;
stream S2 segment. J Am Soc Nephrol. 2011;22(8):1505-1516. 192(2):142-150.
22. Nguyen HT, Woodard JC. Intranephronic calculosis in rats: an 39. Swaminathan S, Rosner MH, Okusa MD. Emerging therapeutic
ultrastructural study. Am J Pathol. 1980;100(1):39-56. targets of sepsis-associated acute kidney injury. Semin Nephrol.
23. Keenan KP, Coleman JB, Mccoy CL, Hoe CM, Soper KA, Lar- 2015;35(1):38-54.
oque P. Chronic nephropathy in ad libitum overfed Sprague- 40. Darisipudi MN, Thomasova D, Mulay SR, et al. Uromodulin
Dawley rats and its early attenuation by increasing degrees of triggers IL 1b dependent innate immunity via the NLRP3 inflam-
dietary (caloric) restriction to control growth. Toxicol Pathol. masome. J Am Soc Nephrol. 2012;23(11):1783-1789.
2000;28(6):788-798. 41. Kurts C, Panzer U, Anders HJ, Rees AJ. The immune system and
24. Radi ZA, Stewart ZS, O’Neil SP. Accidental and programed cell kidney disease: basic concepts and clinical implications. Nat Rev
death in investigative and toxicologic pathology. Curr Protoc Immunol. 2013;13(10):738.
Toxicol. 2018;76(1):e51. 42. Lopez-Novoa JM, Quiros Y, Vicente L, Morales AI, Lopez-Hernan-
25. Radi ZA, Vogel WM, LaBranche T, et al. Renal and hematologic dez FJ. New insights into the mechanism of aminoglycoside nephro-
comparative effects of dissociated agonist of the glucocorticoid toxicity: an integrative point of view. Kid Int. 2011;79(1):33-45.
receptor and prednisone in dogs with and without food restriction. 43. Naesens M, Kuypers DR, Sarwal M. Calcineurin inhibitor
Int J Toxicol. 2018;37(3):223-233. nephrotoxicity. Clin J Am Soc Nephrol. 2009;4(2):481-508.
Radi 13

44. Henry SP, Johnson M, Zanardi TA, et al. Renal uptake and toler- 48. Radi ZA, Stewart ZS, Grzemski FA, Bobrowski W. Renal patho-
ability of a 20 -O-methoxyethyl modified antisense oligonucleotide physiologic role of cortical tubular inclusion bodies. Toxicol
(ISIS 113715) in monkey. Toxicology. 2012;301(1-3):13-20. Pathol. 2013;41(1):32-37.
45. Van Vleet TR, Schnellmann RG. Toxic nephropathy: environ- 49. Yang T, Michele DE, Park J, et al. Expression of peroxisomal
mental chemicals. Sem Nephrol. 2003;23(5):500-508. proliferator-activated receptors and retinoid X receptors in the
46. Levin S, Friedman RM, Cortez E, et al. Lesions and identification kidney. Am J Physiol. 1999;277(6):966-973.
of crystalline precipitates of glycoprotein IIb-IIIa antagonists in 50. Stefanini S, Nardacci R, Farioli-Vecchioli S, Pajalunga D, Sartori
the rat kidney. Toxicol Pathol. 1999;27(1):38-43. C. Liver and kidney peroxisomes in lactating rats and their pups
47. Herzyk DJ, Bugelski PJ, Hart TK, Wier PJ. Preclinical safety of after treatment with ciprofibrate. Biochemical and morphometric
recombinant human interleukin-18. Toxicol Pathol. 2003;31(5): analysis. Cell Mol Biol (Noisy-le-Grand, France). 1999;45(6):
554-561. 815-829.

You might also like