ChapterPDF AvailableLiterature Review

Animal Models and Renal Biomarkers of Diabetic Nephropathy

Authors:

Abstract and Figures

Diabetes mellitus (DM) is the first cause of end stage chronic kidney disease (CKD). Animal models of the disease can shed light on the pathogenesis of the diabetic nephropathy (DN) and novel and earlier biomarkers of the condition may help to improve diagnosis and prognosis. This review summarizes the most important features of animal models used in the study of DN and updates the most recent progress in biomarker research.
Content may be subject to copyright.
Adv Exp Med Biol - Advances in Internal Medicine
https://doi.org/10.1007/5584_2020_527
#Springer Nature Switzerland AG 2020
Animal Models and Renal Biomarkers
of Diabetic Nephropathy
Laura Pérez-López, Mauro Boronat, Carlos Melián,
Yeray Brito-Casillas, and Ana M. Wägner
Abstract
Diabetes mellitus (DM) is the rst cause of end
stage chronic kidney disease (CKD). Animal
models of the disease can shed light on the
pathogenesis of the diabetic nephropathy
(DN) and novel and earlier biomarkers of the
condition may help to improve diagnosis and
prognosis. This review summarizes the most
important features of animal models used in
the study of DN and updates the most recent
progress in biomarker research.
Keywords
Animal models · Chronic kidney disease ·
Creatinine · Cystatin C · Diabetes mellitus ·
Diabetic nephropathy · Early markers ·
Glomerular ltration rate · Kidney injury
molecule-1 · Obesity · Symmetric
dimethylarginine
1 Introduction
Diabetic nephropathy (DN) is a common compli-
cation of diabetes mellitus (DM) occurring in
2040% of people with diabetes (Dronavalli
et al. 2008). Although cardiovascular diseases
are the rst cause of death, approximately
1020% of people with DM die because of kid-
ney failure, and DM is considered the rst cause
of end stage chronic kidney disease (CKD)
(World Health Organization (WHO) 2019).
Improved diagnosis and treatment of renal dis-
ease has led to better prognosis (Andrésdóttir
et al. 2014). Furthermore, detecting the disease
at an earlier stage and building on our understand-
ing of the mechanisms of the disease may help to
improve diagnosis further.
Recently, many reports have proposed a wide
number of markers of CKD, and it has been
shown that many of them reect damage of one
specic part of the nephron (Colhoun and
Marcovecchio 2018; Domingos et al. 2016; Kim
et al. 2013; Kem et al. 2010; Carlsson et al. 2017;
L. Pérez-López and Y. Brito-Casillas
Institute of Biomedical and Health Research (IUIBS),
University of Las Palmas de Gran Canaria (ULPGC), Las
Palmas de Gran Canaria, Spain
M. Boronat and A. M. Wägner (*)
Institute of Biomedical and Health Research (IUIBS),
University of Las Palmas de Gran Canaria (ULPGC), Las
Palmas de Gran Canaria, Spain
Department of Endocrinology and Nutrition, Complejo
Hospitalario Universitario Insular Materno-Infantil, Las
Palmas de Gran Canaria, Spain
e-mail: ana.wagner@ulpgc.es
C. Melián
Institute of Biomedical and Health Research (IUIBS),
University of Las Palmas de Gran Canaria (ULPGC), Las
Palmas de Gran Canaria, Spain
Department of Animal Pathology, Veterinary Faculty,
University of Las Palmas de Gran Canaria, Las Palmas de
Gran Canaria, Arucas, Las Palmas, Spain
Colombo et al. 2019a). Albuminuria and
microalbuminuria have traditionally been consid-
ered as markers of glomerular damage, and they
have also been considered the rst alterations that
can be detected in DN (American Diabetes Asso-
ciation 2004). However, recent studies have
shown that some patients with DM have CKD
in the absence of microalbuminuria (MacIsaac
et al. 2004; Lamacchia et al. 2018; Nauta et al.
2011; Zeni et al. 2017). In addition, the renal
tubule could also play an important role in the
development of DN (Colombo et al. 2019a). In
fact, proteinuria mainly occurs after increased
glomerular capillary permeability, but it is also a
result of impaired reabsorption by the epithelial
cells of the proximal tubule (DAmico and Bazzi
2003). Thus, the use of tubular markers could also
be benecial for the diagnosis of DN. Indeed, the
search for new renal biomarkers could lead to an
earlier detection of renal damage. Likewise, ani-
mal models are important for improved under-
standing of the development and progression of
DN. This review updates the most recent progress
in biomarker research and summarizes the most
important features of animal models used in the
study of DN.
2 Brief Review of Kidney
Anatomy and Physiology
The main functions of the kidney are ltration and
excretion of metabolic waste products from the
bloodstream, regulation of electrolytes, acidity
and blood volume, and contribution to blood
cell production (Rayner et al. 2016).
The nephron is the functional unit of the kid-
ney. Each nephron is formed by a glomerulus, a
proximal convoluted tubule, loop of Henle, and
distal convoluted tubule. The last part of the
nephron is the common collecting duct, and is
shared by many nephrons (Rayner et al. 2016;
National Institute of Diabetes and Digestive and
Kidney Disaese (NIDDK) 2019).
The glomerulus is the ltering unit of the
nephron. Within the glomerulus, the podocytes
are specialized cells lining the outer surfaces of
the bed of capillaries, which have interdigitated
foot processes that play an important role in the
process of ltration. In fact, podocyte damage
leads to proteinuria (Pavenstädt 2000) Podocytes
are part of the glomerular barrier, and protein
barrier passage of a normal kidney is mainly
composed by low molecular weight protein.
However, after its structural integrity is affected,
high molecular weight proteins can pass through
the glomerulus (DAmico and Bazzi 2003).
In regard to the anatomy of the glomerulus, it
has two poles: (1) a vascular pole with the afferent
and the efferent arterioles; and (2) a urinary pole
with the exit to the proximal tubule. The blood is
ltered in a specialized capillary network through
the glomerular barrier, which yields the ltrated
substances into Bowmans capsule space, and
then into the renal tubules. The glomerular barrier
is composed by ve layers: (1) the inner layer is
the glycocalyx covering the surface of the endo-
thelial cells; (2) the fenestrated endothelium,
(3) the glomerular basement membrane, (4) the
slit diaphragm between the foot-processes of the
podocytes; and (5) the sub-podocyte space
between the slit diaphragm and the podocyte
cell body (Rayner et al. 2016).
Another important structure for the process of
ltration and for the regulation of blood pressure
is the juxtaglomerular apparatus, which is adja-
cent to the glomerulus. This structure is formed
by the macula densa (cells inside the cortical of
the thick ascending limb of Henle), mesangial
cells, and the terminal parts of the afferent arteri-
ole that include renin-producing cells. Release of
renin is stimulated by decreased sodium concen-
tration in the macula densa, systemic volume loss
or reduced blood pressure (Peti-Peterdi and Harris
2010; Castrop and Schießl 2014).
3 Pathophysiology of Diabetic
Nephropathy
Hyperglycemia, hypertension and obesity are
considered risk factors for DN (Mogensen et al.
1983; Câmara et al. 2017; Kanasaki et al. 2013).
Therefore, DN is a heterogeneous syndrome that
is common in people with type 1 (T1) and type
2 (T2) DM, although in patients with T2DM and
L. Pérez-López et al.
metabolic syndrome, more heterogeneous
mechanisms are involved. DN has been classi-
cally considered a process with the following
sequence of disorders: glomerular hyperltration,
progression of albuminuria, decline of glomerular
ltration rate (GFR) and, nally end stage renal
disease (Mogensen et al. 1983); and different
pathways that involve hemodynamic, metabolic
and inammatory factors play a role in its devel-
opment (Table 1).
3.1 Hemodynamic Factors
Glomerular hyperltration is considered an alter-
ation of early DN and it has been identied in
1040% of people with early T1DM, and around
40% of patients with T2DM (Mogensen et al. 1983;
Premaratne et al. 2015). However, its role as a
leading cause of DN needs further research. In addi-
tion, the hyperltration mechanism is not well
understood yet: it could be the result of a combina-
tion of hemodynamic, vasoactive, and tubular
factors (Dronavalli et al. 2008;Zenietal.2017).
Moreover, inhibition of tubuloglomerular feedback
could be the main mechanism involved (Zeni et al.
2017). Persistent hyperglycemia produces tubular
growth and increased tubular sodium reabsorption,
and reduces the delivery of sodium to the macula
densa, eliciting the release of renin and the activation
of the renin-angiotensin system (RAS). This leads to
an inhibition of the tubuloglomerular feedback,
causing vasodilation of the afferent arteriole, which
increases single-nephron glomerular ltration rate
and consequently leads to hyperltration
(Premaratne et al. 2015; Vallon and Thomson
2012). An implication of the sodium-glucose
co-transporter 2 (SGLT2) in this process has been
suggested. SGLT2s are expressed in the proximal
tubule and their function is the reuptake of glucose
and sodium (ratio 1:1), thus their activity is
stimulated by the increased glucose ltration in dia-
betic subjects (Zeni et al. 2017; Premaratne et al.
2015; Hans-Joachim et al. 2016). Vasodilating
substances, such as nitric oxide and cyclo-
oxygenase 2 derived prostanoids, also take part in
the hyperltration process (Wolf et al. 2005).
Whether they play a key or secondary role in the
pathogenesis of hyperltration is not well dened.
This process, which is associated with increased
intraglomerular pressure, could lead to podocyte
stress and nephron loss (Hans-Joachim et al.
2016). Additionally, obesity has been considered
as an independent risk factor for CKD, and
hyperltration has also been detected in
non-diabetic obese subjects. In addition, the kidney
produces components of the RAS that specically
constrain the efferent rather than the afferent arteri-
ole, increasing GFR and glomerular pressure
(Yacoub and Campbell 2015). Several studies
have demonstrated local production of RAS
components in adipose tissue (Sharma and Engeli
2006; Giacchetti et al. 2002) and it has been
suggested that RAS could be overactivated in
patients with obesity (Sharma and Engeli 2006;Xu
et al. 2017). Indeed, a decrease in RAS activity has
been reported in obese women after weight loss
(Engeli et al. 2005).
Table 1 Main factors involved in the development of diabetic nephropathy
Mechanisms involved in the pathogenesis of diabetic nephropathy
Hemodynamic factors Metabolic and inammatory factors
Vasodilation of the afferent arteriole of
the glomerulus
Increased formation of advanced glycation end-products
Increased glomerular ltration rate Podocyte stress
Implication of RAS, nitric oxide and
cyclo-oxygenase 2
TGF-β1 is a probrotic cytokine that plays an essential role in inammatory
and brotic processes
Increased glomerular pressure Obesity leads to increased leptin concentration, decreased adiponectin
concentration, and increased cytokine production
Abbreviations: RAS renin angiotensin system, TGF-β1transforming growth factor β1
Animal Models and Renal Biomarkers of Diabetic Nephropathy
3.2 Metabolic and Inflammatory
Factors
Formation of advanced glycation end-products
(AGE), resulting from the reduction of sugars, is
increased during chronic hyperglycemia. Accu-
mulation of AGE appears to stimulate production
of cytokines and renal brosis (Forbes and Coo-
per 2007). Inltration by inammatory cells
(monocytes, macrophages and lymphocytes)
precedes brosis, and these inammatory cells
are responsible for the production of reactive
oxygen species, inammatory cytokines and
probrotic cytokines (Kanasaki et al. 2013).
Among the latter, transforming growth factor
beta 1 (TGF-β1) plays an essential role in inam-
mation and brosis, and together with other
cytokines such as connective tissue growth factor,
platelet-derived growth factor and broblast
growth factor 2; TGF-β1 is involved in broblast
activation (Kanasaki et al. 2013). This produces
an increased deposition of extracellular matrix in
the interstitial space, as well as glomerular base-
ment membrane thickening, which could lead to
podocyte apoptosis and, as a consequence, to
increased vascular permeability in the glomerulus
(Wolf et al. 2005).
Additionally, in obese people, high leptin and
low adiponectin concentrations result in an
increased secretion of several adipokines
(i.e. tumor necrosis factor-a, interleukin-6,
interleukin-18) that promote an inammatory state,
also leading to extracellular matrix accumulation
and renal brosis (Câmara et al. 2017; Straczkowski
et al. 2007;Kernetal.2001). In obese mice, reduced
plasma adiponectin contributes to albuminuria and
podocyte disfunction (Sharma et al. 2008).
3.3 Histologic Changes
The main histologic changes observed in kidneys
of people with diabetes are located in the glomer-
ulus, and include glomerular sclerosis
(Kimmelstiel-Wilson nodules), basement mem-
brane thickening and mesangial expansion.
Tubulo-interstitial and arteriolar lesions have
been commonly described as late lesions in
patients with T1DM. Nevertheless, some patients
with T2DM can show tubulo-interstitial and/or
arteriolar lesions with preserved glomerular struc-
ture (Fioretto and Mauer 2007).
4 Animal Models
4.1 Rodents
4.1.1 Mouse Models
Rodents are the most studied animal models of
human DN. They can show spontaneous or
induced diabetes (Kachapati et al. 2012; Song
et al. 2009), and DN might develop in the course
of the disease, or by induction of renal damage by
unilateral nephrectomy, or ischemia and reperfu-
sion (Song et al. 2009; Kitada et al. 2016). How-
ever, an animal model that develops all of the
features of DN is not available (Betz and Conway
2014). Based on the clinical characteristics of DN
in humans, the Animal Models of Diabetic
Complications Consortium (AMDCC) has
published some criteria to dene acceptable
models of renal disease in mice with diabetes:
(1) 50% decline in GFR; (2) 100 fold increase in
proteinuria in comparison to matched controls of
the same strain, age and gender; (3) presence of
pathologic alterations such as mesangial sclerosis,
arterial hyalinosis, 50% thickening of the glomer-
ular basement membrane or tubulointerstitial
brosis. Nonetheless, to date, no animal model
fullls all these criteria (Diabetes Complications
Consortium (DiaComp) 2003).
T1DM rodent models include streptozotocin
induced DM or spontaneous models due to genetic
mutations, such as AKITA and OVE26 mice.
T2DM genetic models are leptin decient (ob/ob
mice) or have inactivating mutations in the leptin
receptor (db/db mice) (Kitada et al. 2016;Alpersand
Hudkins 2011), but there are also models of induced
T2DM, usually through a high fat diet (De Francesco
et al. 2019; Ingvorsen et al. 2017). Hypertension
plays an important role in the progression of
human DN, and the deciency of endothelial nitric
oxide synthase (eNOS) through knockout of eNOS
genes, has led to accelerated renal damage in db/db
L. Pérez-López et al.
and streptozotocin-treated mice. eNOS
/
C57BL/
J
db
mice can develop T2DM, obesity, hypertension,
albuminuria, marked mesangial expansion and
mesangiolysis (Nakagawa et al. 2007).
Recently, the black and tan, brachyury (BTBR
obese) mouse strain, which is spontaneously insu-
lin resistant, has gained importance in the eld of
the study of DN. This strain with ob/ob leptin
deciency mutation on BTBR mouse back-
ground, has been considered one of the best
models of human DN because it rapidly develops
pathological changes seen in human DN, such as
increased glomerular basement membrane thick-
ness, mesangial sclerosis, focal arteriolar
hyalinosis, mesangiolysis, mild interstitial bro-
sis and podocyte loss (Hudkins et al. 2010).
4.1.2 Rat Models
The most studied rat models are the Zucker dia-
betic fatty (ZDF-fa/fa) rat and the Wistar fatty rat
(Kitada et al. 2016; Hoshi et al. 2002). Both have
an autosomal recessive mutation in the fa gene
that encodes the leptin receptor, and both are
crossbred with the insulin resistant Wistar Kyoto
rats. The ZDF-fa/fa and Wistar fatty rats can
develop albuminuria and renal alterations, such
as tubular cell damage and tubulointerstitial bro-
sis, although nodular glomerular lesions or
mesangiolysis have not been observed (Kitada
et al. 2016; Hoshi et al. 2002). Another rat strain
that is considered a model of DN is The Otsuka
Long-Evans Tokushima Fatty (OLETF), in which
multiple recessive genes are involved in the
development of DM. These rats exhibit mild obe-
sity, hyperinsulinemia with late onset of
hyperglycaemia, and can present mesangial
matrix expansion, glomerulosclerosis and tubular
cell damage (Kawano et al. 1994,1992).
4.2 Companion Animals
These animals have a great interest as animal
models since they develop spontaneous DM and
share the human environment (Brito-Casillas
et al. 2016). However, it is unclear if dogs and
cats with diabetes develop DN.
4.2.1 Cats
Around 80% of cats with DM have T2DM (Nel-
son and Reusch 2014). As in humans, obesity is a
risk factor for DM, which also represents a prob-
lem of an epidemic proportion since approxi-
mately 3550% of domestic cats are overweight
or obese (Hoenig 2012). Obese cats also show
some of the disorders observed in people with the
metabolic syndrome; they can present insulin
resistance and higher concentrations of very
low-density lipoproteins (VLDL), triglycerides
and cholesterol (Hoenig 2012; Jordan et al.
2008). Despite these alterations, hypertension
has not been linked to feline obesity or DM, and
atherosclerosis has not been described either
(Jordan et al. 2008; Payne et al. 2017). Lower
concentration of angiotensin-converting enzyme
2 has been found in the subcutaneous adipose
tissue of overweight or obese cats compared to
those with a low body condition score, but
whether this could represents a negative mecha-
nism against blood pressure is unknown (Riedel
et al. 2006).
Additionally, the link between DM and renal
disease is unclear in cats, and discrepancies can
be found in the literature. In two retrospective,
epidemiological studies, no relationship was
found between both diseases (Greene et al.
2014; Barlett et al. 2010). In contrast, higher
prevalences of proteinuria and microalbuminuria
have been detected in cats with DM compared to
age matched controls (Al-Ghazlat et al. 2011)
and, recently, in a retrospective study of a popu-
lation of 561 adult cats, age-adjusted multivariate
regression analysis showed an association
between both diseases (Pérez-López et al. 2019).
Thus, the cat could be a useful model of DN,
although prospective studies are still needed for
a better understanding and evaluation of the asso-
ciation between feline DM and renal disease.
4.2.2 Dogs
Obesity in dogs is able to induce
hyperinsulinemia, to increase blood pressure and
to produce glomerular hyperltration. Dogs with
obesity can develop structural changes in the kid-
ney, such as glomerular basal membrane
Animal Models and Renal Biomarkers of Diabetic Nephropathy
thickening and mesangial expansion (Henegar
et al. 2001). Insulin sensitivity decreases around
35% in obese dogs. However, obesity does not
cause T2DM in these animals (Chandler et al.
2017). Indeed, T1DM is the most commonly
recognized form of DM in dogs, although they
can also develop DM secondarily to dioestrus,
Cushings syndrome or medications (Nelson and
Reusch 2014). Regarding the relationship
between DM and CKD, it has not been fully
investigated in dogs. However, some studies
have shown that markers of vascular resistance
(ultrasound renal resistive index and pulsatility
index), which are associated with progression of
kidney disease and hypertension in humans, are
also positively correlated with glycemic status in
dogs (Priyanka et al. 2018; Novellas et al. 2010).
In a case-control and age-matched study, dogs
with alloxan-induced DM, uninephrectomized
4 weeks after the induction of DM, were proposed
as a valuable model for the study of DN, since
they developed greater glomerular basement
membrane thickening and mesangial expansion,
compared to controls, already 1 year after DM
induction (Steffes et al. 1982).
4.3 Production Animals
Mainly two species could represent this group as
animal models of DN: rabbits and swine. How-
ever, few studies have been reported on rabbits,
and they have been mainly focused on the role of
obesity in kidney function (Dwyer et al. 2000;
Antic et al. 1999). One study showed an increased
hyaluronean content in the renal medulla of
rabbits with induced high-fat diet induced obesity
(Dwyer et al. 2000). Higher renal medullary
hyaluronean content has also been reported in
humans and rodent models with DM or with
renal alterations (Stridh et al. 2012). In regard to
the swine, it is considered a more valuable animal
model, since it closely resembles human anatomy
and physiology (Zhang and Lerman 2016;Li
et al. 2011; Rodríguez-Rodríguez et al. 2020;
Spurlock and Gabler 2008). Metabolic syndrome
can be induced through high fat diet in the
Ossabaw and Iberian swine. These pigs exhibit
obesity, insulin resistance, hypertension and
dyslipidemia. Regarding renal disease, kidney
hypertrophy, increased GFR, renal tubular bro-
sis and renal adiposity have all been reported
(Zhang and Lerman 2016; Li et al. 2011;
Rodríguez-Rodríguez et al. 2020).
Further information in relation to animal
models can be found in Table 2.
5 Assessment of Renal Function
Criteria to diagnose CKD are well established in
human medicine. However, there are a large num-
ber of markers, many still under investigation,
that could allow an early detection of impaired
renal function. The assortment of markers also
represents a variety of mechanisms involved in
kidney injury, reecting damage of different parts
of the nephron as well (Fig. 1).
5.1 Routine Evaluation of DN
and Gold Standard Methods
In humans, CKD is dened as abnormalities of
kidney structure or function, present for more
than 3 months, and with implications for health.
Patients with altered kidney function present
albuminuria and/or a decline in GFR (Levin
et al. 2013).
Human DN is diagnosed when urinary albu-
min to creatinine ratio (ACR) is above 30 mg/g,
although some guidelines recommend the use of
different ACR thresholds for men (>25 mg/g) and
women (>35 mg/g). Despite ACR being the main
tool used to diagnose DN, some patients with
either T1 or T2DM who have decreased GFR do
not show elevated ACR (Gross et al. 2005;
Caramori et al. 2003). Therefore, GFR should
also be measured in patients with DM to rule
out CKD. In fact, estimation of GFR is considered
the routine method to evaluate renal function in
patients with CKD. The most recent guidelines
recommend the interpretation of both albuminuria
and GFR for the diagnosis and staging of CKD
(Levin et al. 2013).
L. Pérez-López et al.
Table 2 Animal models of diabetic nephropathy
Animal models of induced diabetes with development of DN
Animal models of spontaneous diabetes
with development of DN
Spontaneous animal model of diabetes and unclear
development of DN
Animal model Advantages Disadvantages
Animal
model Advantages Disadvantages
Animal
model Advantages Disadvantages
Models
of
T1DM
Streptozotocin
treated
C57BL/6
mouse
The most studied
strain
Less susceptibility to
develop renal injury
than other strains
AKITA
mouse.
Early
pathological
changes of DN
Strain-
dependent
susceptibility
Diabetic
dog
Gene-environment
interaction
Development
of DN is still
unclear
Easy to breed and
long life span
Environmental
factors shared with
humans
Streptozotocin
treated DBA/2
mouse
More susceptible to
renal injury than
C57BL/6 mouse
Tubulointerstitial
brosis does not occur
OVE26
mouse
To study
advanced DN
Poor viability
Models
of
T2DM
C57BL/6
mouse on high
fat diet
Develops
metabolic
syndrome
(increased SBP and
lipids)
Inter-individual
phenotypic variability
of the amount of food
intake after receiving a
high fat diet
eNOS
/
/db/
db
mouse
Development
of glomerular
lesions
characteristic
of advanced
DN
Few studies,
possible
difculties
with breeding
Diabetic
cat
Similar mechanisms
of human T2DM,
with environmental
factors and polygenic
interactions
Development
of DN is still
unclear
Renal injury could
be due to renal
lipid accumulation
Phenotype more
pronounced in males
Zucker
diabetic fatty
(ZDF-fa/fa) rat
Development of
tubulointerstitial
brosis
Does not develop
features of advanced
DN
BTBR
ob/ob
mice
Rapidly
resembles
alterations of
advanced
human DN
Modest
interstitial
brosis
Ossabaw pig
on high fat diet
Develops
metabolic
syndrome,
increased GFR,
renal tubular
brosis, renal
adiposity
Expensive and
specialized
husbandry
Abbreviations: IgG immunoglobulin G; kidney injury molecule -1, NGAl neutrophil gelatinase-associated lipocalin, SDMA symmetric dimethylarginine, VEGF vascular
endothelial growth factor, RBP4 retinol binding protein 4, suPAR soluble urokinase type plasminogen activator receptor, TGFβ1transforming growth factor β-1
Animal Models and Renal Biomarkers of Diabetic Nephropathy
The gold standard for assessing GFR is the
plasma or urinary clearance of an exogenous l-
tration marker, such as inulin (Stevens and Levey
2009), Cr-EDTA,
125
I-iothalamate or iohexol.
However, direct GFR measurement is difcult to
perform and is time-consuming; it requires the
injection of a suitable marker and several urine
sample collections (Stevens and Levey 2009;
Levey et al. 2003). Alternatively, equations for
estimation of GFR, generally based on serum
creatinine levels, are used in clinical practice,
and their use is recommended in conjunction
with other markers of renal function, such as
cystatin C (Levin et al. 2013). However, early
alterations of DN are usually associated with
hyperltration, and these equations are less pre-
cise at higher values of GFR, and tend to under-
estimate GFR in the hyperltration state (Levin
et al. 2013; Tuttle et al. 2014). Among available
formulas, the Chronic Kidney Disease Epidemi-
ology Collaboration (CKD-EPI) has been
suggested to be the best one to evaluate early
renal impairment in patients with DM,
normoalbuminuria and hyperltration (Lovrenčić
et al. 2012).
.
Recent studies suggest that the
CKD-EPI cystatin C based equations for estima-
tion of GFR, are those that best t the GFR
measurement in patients with DM. However, in
general, it is considered that GFR equations show
high variability in people with DM (Cheuiche
et al. 2019).
As stated above, the development of DN is
unclear in companion animals, but diagnosis of
CKD is based on serum creatinine or symmetric
dimethylarginine (SDMA) concentration together
with urinary protein-creatinine ratio (International
Renal Interest Society (IRIS) 2015a,2017), and
there is still a lack of standardization in the
methods of GFR measurement and their interpre-
tation (International Renal Interest Society (IRIS)
2015b). Only one study proposed a method to
estimate GFR in cats. It was based on serum
creatinine and it was adjusted for a marker of
muscle mass. However, this formula did not
prove to be a reliable estimation of GFR (Finch
et al. 2018).
Glomerular filtration:
Creatinine
SDMA
Cystatin C
Glomerular barrier:
IgG
Albumin
Proximal tubule:
Cystatin C
RBP4
KIM-1
Thick ascending
limb of Henle’s
loop and distal
tubule:
Uromodulin
NGAL
Collecting duct:
NGAL
Inflammation and fibrosis:
TGFβ1
VEGF
suPAR
Fig. 1 Classication of markers of renal damage according to part of the nephron involved
L. Pérez-López et al.
5.2 Indirect Markers of Glomerular
Filtration Rate
5.2.1 Serum Creatinine
Creatinine is a break-down product of creatine
phosphate in muscle tissue (113 Daltons). It is
not metabolized and is entirely cleared by the
kidneys with minimal reabsorption by the renal
tubules (Ferguson and Waikar 2012; Kavarikova
2018). Thus, blood creatinine concentration
increases when GFR declines. However, in the
early stage of CKD, subtle changes in GFR do not
alter creatinine concentration. In humans, other
markers seems to correlate better with GFR than
creatinine (El-khoury et al. 2016) and, in dogs, its
concentrations increase only when around 50% of
renal function has been lost (Hokamp and Nabity
2016; Nabity et al. 2015). In addition, both in
humans and companion animals, creatinine con-
centration depends on lean body mass, and it has
high inter-individual variability (Delanaye et al.
2017; Hall et al. 2014a,2015). Other factors, such
as hydration and blood volume status, urinary
obstructions or urinary infections, can also affect
creatinine concentration (Hokamp and Nabity
2016; Blantz 1998).
5.2.2 Symmetric Dimethylarginine
SDMA is a catabolic product of arginine-
methylated proteins (202 daltons), which is
mainly excreted through the kidneys and is not
reabsorbed or secreted by the tubules (Nabity
et al. 2015; McDermott 1976). Thus, SDMA con-
centration is affected by GFR in inverse linear
relationship in humans and in companion animals
(Pelander et al. 2019; Hall et al. 2014b; Relford
et al. 2016). Studies in companion animals have
shown that, in contrast to creatinine, SDMA is not
affected by muscle mass (International Renal
Interest Society (IRIS) 2015a,2017). On the
other hand, human studies have found no correla-
tion or an inverse correlation between SDMA and
body mass index (Schepers et al. 2011;
Schewedhelm et al. 2011; Potočnjak et al.
2018). Furthermore, discrepancies in the levels
of SDMA in patients with DM have been reported
among different studies. The offspring cohort
study from the Framingham Heart Study did not
nd a relationship between SDMA and insulin
resistance (Schewedhelm et al. 2011). Moreover,
an inverse correlation between SDMA and
glycosylated hemoglobin or fructosamine levels
has been reported in T2DM patients, i.e. those
with poor glycemic control had lower SDMA
concentration (Can et al. 2011). In another
study, SDMA was increased in patients with
T2DM and microalbuminuria, and this marker
predicted impaired renal function and cardiovas-
cular disease in this population (Zobel et al.
2017). Additionally, SDMA has been positively
associated with proteinuria and inversely
associated with GFR in patients with CKD
and T2DM, and SDMA to asymmetric
dimethylarginine (ADMA) ratio was one of the
strongest predictive markers of renal function
decline (Looker et al. 2015). ADMA is another
byproduct of the proteolytic breakdown of
nuclear proteins. It is an inhibitor of NOS and is
considered to play a role in endothelial dysfunc-
tion (Sibal et al. 2010).
Interestingly, another study showed that young
patients with T1DM and microalbuminuria and
high GFR, had lower SDMA concentrations com-
pared to normoalbuminuric T1DM patients; and
over time, microalbuminuric patients showed an
increment in SDMA concentration, probably
reecting a decline in GFR (Marcovecchio et al.
2010). Therefore, the lower levels of SDMA
observed in patients with DM might be explained
by hyperltration occurring in the early stages of
DN. However, this requires further investigation,
as other explanations have been suggested (Zsuga
et al. 2007; Closs et al. 1997; Simmons et al.
1996; Siroen et al. 2005; Nijveldt et al. 2003).
In cats with DM, lower levels of SDMA have
also been observed. However, in this species,
further research on the relationship between DM
and CKD is still needed (Langhorn et al. 2018).
In relation to the inverse association observed
between SDMA and GFR, one study in humans,
comparing SDMA to the renal clearance of an
exogenous molecule (
125
Isodium iothalamate),
showed that SDMA had a stronger inverse corre-
lation with GFR than creatinine (El-khoury et al.
2016). In contrast, in dogs, the inverse correlation
Animal Models and Renal Biomarkers of Diabetic Nephropathy
between SDMA and GFR determined through
iohexol clearance was similar to the correlation
observed between creatinine and GFR. However,
SDMA was considered an earlier marker of GFR
than creatinine, as SDMA was able to detect a
decrease in renal function <20% on average,
whereas creatinine increased when renal function
was reduced by 50% (Hall et al. 2016). Similarly,
in cats, the inverse correlation between SDMA
and GFR measured by iohexol clearance was
similar to the correlation observed between creat-
inine and GFR, but the sensitivity to detect
impaired renal function was higher using SDMA
(100 vs 17%) (Hall et al. 2014b). The established
International Renal Interest Society guidelines
consider that SDMA persistently >14 ng/ml is
consistent with CKD (International Renal Interest
Society (IRIS) 2015a). This cut-off (>14 ng/ml)
of SDMA is able to detect a decrease of 24% from
the median of GFR established for healthy cats,
with a sensitivity of 91% (Hall et al. 2014b).
5.3 Markers of Glomerular Damage
5.3.1 Urine Albumin
Albumin is an intermediate molecular weight pro-
tein (69 KD) and its urinary concentration could
increase with moderate disorders affecting the
permeability of the glomerular barrier (DAmico
and Bazzi 2003). The initial alterations of the
glomerular barrier usually involve a loss of
restriction to passage of negatively charged
proteins (especially albumin). Albuminuria can
also occur when the tubular cells are damaged
and tubular reabsorption is impaired, although
intense albuminuria is usually glomerular in ori-
gin (Nauta et al. 2011;DAmico and Bazzi 2003).
The evaluation of a single albumin measurement
is not recommended because it can vary during
the day. Therefore, the measurement of urinary
ACR in a random or rst morning sample, or
through a 24 h urine collection, with a measure-
ment of creatinine, is advised (Basi et al. 2008). In
addition, diabetic rodent models that develop
albuminuria are considered particularly useful
for the study of human DN (Diabetes
Complications Consortium (DiaComp) 2003). In
rats, microalbuminuria has been associated with
reduced albumin reabsorption in the proximal
tubule in early DN (Tojo et al. 2001). Some
studies have used ACR to study renal function
in dogs (Tvari-jonaviciute 2013). However, in
clinical practice of small domestic animals, the
protein creatinine ratio is used instead of albumin.
Although clinical interpretation of albuminuria is
not well established in dogs and cats, borderline
protein-creatinine ratio between 0.2 and 0.5 mg/
mg in dogs, and between 0.2 and 0.4 mg/mg in
cats, are suggestive of microalbuminuria, and its
monitorization is recommended (International
Renal Interest Society (IRIS) 2017). It should
also be highlighted that albuminuria is not spe-
cic for kidney function and it could appear in the
presence of non-renal diseases (i.e., hyperadreno-
corticism, urinary tract infections or neoplasms)
(Kivarikova 2015).
5.3.2 Urine Immunoglobulin G
Immunoglobulin (IgG) is a high molecular weight
protein (160 kDa) that is involved in antibody-
mediated immunity. Due to its size, this protein
cannot pass through an intact glomerular barrier.
Therefore, urine detection of IgG reects glomer-
ular damage (DAmico and Bazzi 2003). IgG has
been associated with albuminuria in patients with
DM (Carlsson et al. 2017), but few studies have
investigated IgG in animals. In dogs, urine detec-
tion of IgG has been associated with X-linked
hereditary nephropathy even before the onset of
proteinuria. The authors hypothesized that in
dogs some degree of alteration of the glomerular
basement membrane could allow the passage of
proteins not detected by the usual assays to mea-
sure proteinuria (Nabity et al. 2012). Similarly, in
humans, IgG has been detected in normo-
albuminuric patients with T2DM, and it has
been considered a predictive marker of albumin-
uria (Narita et al. 2006).
5.4 Markers of Tubular Damage
5.4.1 Serum and Urine Cystatin C
Cystatin C is a low molecular weight protein
(13 kDa) that is considered as a marker of both
L. Pérez-López et al.
GFR and proximal tubular damage. Cystatin C is
freely ltered by the glomerulus, and it is almost
entirely reabsorbed in the proximal tubule by
megalin-mediated endocytosis (Mussap and
Plebani 2004; Kaseda et al. 2007). Its inverse
correlation with GFR is better than that observed
between creatinine and GFR (El-khoury et al.
2016). In addition, studies in animal models
have suggested that cystatin C could reect
impaired kidney function earlier than creatinine
(Song et al. 2009; Togashi and Miyamoto 2013).
For example, after ischaemia-reperfusion injury,
partial unilateral nephrectomy and bilateral
nephrectomy, serum cystatin C concentration
increased before creatinine in BALB/c mice
models (Song et al. 2009). Another report
observed that, in comparison to Zucker diabetic
lean rats, Zucker Diabetic Fatty rats showed
elevations of urinary cystatin C concentration,
along with other renal biomarkers of kidney injury
(β2-microglobulin, clusterin, mu-glutathione
S-transferase and kidney injury molecule-1
(KIM-1)), but not of serum creatinine, before the
appearance of kidney histopathological changes
(Togashi and Miyamoto 2013). Additionally, in
this study, the authors observed that immunohisto-
chemical cystatin C expression was predominantly
localized in the proximal tubules of the renal cor-
tex, supporting a role of tubular damage in the
development of kidney injury due to obesity
(Togashi and Miyamoto 2013).
Furthermore, some advantages of cystatin C as
a marker of CKD compared to creatinine should
be highlighted. In humans, it is less affected by
muscle mass than creatinine and it has lower
inter-individual variability (Stevens et al. 2009).
However, it should be taken into account that the
concentration of cystatin C is subjected to
changes in people with thyroid disorders or glu-
cocorticoid treatment (Risch and Huber 2002;
Fricker et al. 2003).
In human medicine, clinical practice
guidelines for the evaluation and management of
CKD recommend the measurement of cystatin C,
especially in those patients in whom estimated
GFR, based on serum creatinine, might be
expected to be less accurate, or in those patients
with early stages of CKD (estimated GFR
between 4559 ml/min/1.73 m
2
), who do not
have other markers of kidney damage (Levin
et al. 2013). Additionally, cystatin C might be a
predictor of impaired renal function in patients
with T2DM and, although its concentration can
reach higher levels in macroalbuminuric patients,
it is independently associated with GFR, and an
increase in serum and urine cystatin C concentra-
tion has been observed in subjects with DM,
normoalbuminuria and decreased GFR (Kim
et al. 2013; Jeon et al. 2011). Thus, cystatin C
might predict DN in the early stages of impaired
renal function in patients with T2DM (Kim et al.
2013; Jeon et al. 2011).
In contrast, in veterinary medicine, the use of
cystatin C does not seem so advantageous. In cats,
serum cystatin C was not found to be a reliable
marker of GFR, and in regard to urinary cystatin C,
one study showed higher concentrations of this
marker in cats with CKD compared to healthy
cats, although, urinary Cystatin C was below the
detection limit of the assay in some of the cats with
CKD (Ghys et al. 2016; Williams and Archer
2016). In addition, one study reported lower
cystatin C concentration in cats with DM com-
pared to healthy cats (Paepe et al. 2015), although
in this species it is not clear whether CKD could
occur secondarily to DM (Greene et al. 2014;
Barlett et al. 2010;Al-Ghazlatetal.2011; Pérez-
López et al. 2019;Zinietal.2014).
In a similar manner, in dogs, serum cystatin C
does not seem to be superior to creatinine, either,
and the sensitivity of cystatin C to detect CKD
has been considered similar to SDMA and creati-
nine, whereas its specicity was lower (Pelander
et al. 2019; Almy et al. 2002; Marynissen et al.
2016).
5.4.2 Retinol-Binding Protein 4 (RBP4)
Retinol-binding protein is a low molecular weight
protein (21 kDa) that acts as the transport protein
for retinol in plasma. It is produced in the liver
and circulates in plasma bound to transthyretin
(TTR), a protein with a molecular weight of
54 kDa that is too large to pass through the
glomerular barrier. However, around 45% of
serum RBP4 circulates freely and can pass
through this barrier, to be then reabsorbed by
Animal Models and Renal Biomarkers of Diabetic Nephropathy
tubular epithelial cells (Christensen et al. 1999).
When tubular damage occurs, reabsorption of
retinol is decreased, with subsequent loss of
RBP4 into the urine (Zeni et al. 2017). Urinary
RBP4 has been considered as a predictive marker
of CKD in humans and of microalbuminuria in
patients with T2DM (Domingos et al. 2016; Park
et al. 2014). However, one study suggested that
serum RBP4 does not seem to be a better marker
than creatinine or cystatin C to detect GFR
impairment (Donadio et al. 2001). On the other
hand, serum retinol has also been considered a
marker of insulin resistance and cardiovascular
risk factors in humans and animal models (Park
et al. 2014; Cabré et al. 2007; Mohapatra et al.
2011; Graham et al. 2006; Yang et al. 2005;
Akbay et al. 2010), and it could be interesting
for the study of early renal alterations in patients
with metabolic syndrome. Mouse models have
demonstrated that transgenic overexpression of
human RBP4, or injection of recombinant
RBP4, leads to insulin resistance (Yang et al.
2005), although a few reports disagree on this
association (Von Eynatten et al. 2007; Henze
et al. 2008).
In dogs and cats, higher urinary RBP4 concen-
tration has been observed in animals with CKD
compared to healthy animals (van Hoek et al.
2008; Chakar et al. 2017).
5.4.3 Uromodulin (Tamm-Horsfall
Protein)
Uromodulin is a 100 kDa protein synthetized by
the epithelial cells of the thick ascending limb of
Henles loop and the distal convoluted tubule
(Hokamp and Nabity 2016). Therefore, in healthy
individuals it is normal to nd uromodulin in
urine samples, whereas in patients with tubular
damage its urinary concentration is low or even
absent (Chakraborty et al. 2004; Steubl et al.
2016). Correlation between uromodulin and
GFR has only been assessed through equations
of estimated GFR, and conicting results have
been reported. Whereas urinary uromodulin is
positively correlated to GFR, the correlation
between GFR and serum uromodulin has been
reported as positive or negative, depending on
the study (Möllsten and Torffvit 2010; Prajczer
et al. 2010; Fedak and Kuźniewski 2016;
Wiromrat et al. 2019). Nonetheless, several
reports have observed that plasma or serum
uromodulin concentrations are lower in patients
with nephropathy. Uromodulin could be an indi-
cator of renal damage in people with and without
DM (Chakraborty et al. 2004; Steubl et al. 2016;
Möllsten and Torffvit 2010). Kidneys of patients
with very low levels of serum and urinary
uromodulin show more tubular atrophy and
decreased concentration is present at the earliest
stages of CKD (Prajczer et al. 2010). However, in
dogs, uromodulin has been considered a progres-
sion marker rather than an early marker of CKD
(Chakar et al. 2017).
Additionally, studies in uromodulin knockout
mice showed that it has a protective role against
urinary tract infections and renal stone formation
(Rampoldi et al. 2011; Bates et al. 2004; Liu et al.
2010). However, in humans, mutations in the
gene encoding uromodulin lead to a rare autoso-
mal dominant disease, which causes tubuloin-
terstitial damage, but these patients do not show
increased rates of urinary tract infections or renal
stone formation (Rampoldi et al. 2011).
5.4.4 Neutrophil Gelatinase-Associated
Lipocalin (NGAL)
NGAL is a low molecular weight protein of
25 kDa belonging to the lipocalin protein super-
family that is produced in the renal tubules (thick
ascending limb of Henles loop, distal tubule, and
collecting duct) after inammation or tissue
injury (Singer et al. 2013). Its concentration may
also be increased in case of impaired proximal
tubular reabsorption (Singer et al. 2013).
In patients with DM, urinary NGAL shows an
inverse correlation with GFR and a positive cor-
relation with albuminuria (Kem et al. 2010;Fu
et al. 2012a; Nielsen et al. 2011; Vijay et al.
2018). In addition, NGAL concentrations are
higher in normoalbuminuric patients with DM,
compared to non-diabetic control subjects,
which might suggest that tubular damage could
be one of the earliest alterations in patients with
DN (Nauta et al. 2011; Vijay et al. 2018). More-
over, higher levels of urinary NAGL have been
observed in T2DM diabetic patients with
L. Pérez-López et al.
glomerular hyperltration compared to T2DM
with normal GFR and control subjects (Fu et al.
2012b).
In addition, NGAL has been proposed as a
useful indicator of acute kidney injury in human
medicine (Singer et al. 2013; Haase et al. 2011)
and most studies in animal models focus on its
utility to detect acute kidney injury. In dogs,
plasma and urinary NGAL are able to distinguish
dogs with CKD from those with acute kidney
injury (Steinbach et al. 2014). In contrast, in cats
with CKD, this marker did not increase until the
cats reached an advanced stage of the disease
(Wang et al. 2017). Additionally, it should be
highlighted that NGAL concentration could be
inuenced by other conditions, such as urinary
tract infections, different types of neoplasms, pre-
eclampsia and obstructive pulmonary disease
(Giasson et al. 2011; Fjaertoft et al. 2005;
Keatings and Barnes 1997; Bolignano et al.
2010).
5.4.5 Kidney Injury Molecule -1 (KIM-1)
KIM-1 is a type 1 transmembrane glycoprotein
(90 kDa) located in the proximal tubules and,
after tubular injury, its concentration rises before
serum creatinine, whereas it is not detected in the
urine of humans or other species without kidney
damage (Moresco et al. 2018). KIM-1 is a well-
known marker of acute kidney injury, and some
studies suggest that it might be a good marker for
CKD since its concentrations are high in humans
with low GFR or albuminuria. Additionally, in
humans with T2DM and normoalbuminuria or
mild albuminuria, an increment of the urinary
concentration of this marker has been observed
(De Carvalho et al. 2016). Its ability to predict
DN requires further investigation since
discrepancies have been observed (Colombo
et al. 2019a; Nauta et al. 2011). Two longitudinal
studies showed that, the use of KIM-1 in T2DM,
together with pro b-type natriuretic peptide or
beta 2 microglobulin, GFR and albuminuria,
seemed to improve prediction of kidney function
decline (Kammer et al. 2019; Colombo et al.
2019b). In contrast, another study reported that
KIM-1 was not associated with albuminuria
(Nauta et al. 2011), and, in a longitudinal, multi-
center study in T1DM, it did not improve the
prediction of progression of DN compared to
albuminuria and estimated GFR, either (Panduru
et al. 2015). In another large study, from an
extensive set of biomarkers, KIM-1 and CD27
antigen combined, were the most important
predictors of DN progression, although their pre-
dictive power did not improve that of historical
estimated GFR and albuminuria. (Colombo et al.
2019a). It should also be highlighted that other
conditions such as sepsis and urinary tract disease
can increase urinary KIM-1 (Moresco et al.
2018).
In animal models, higher concentrations of this
marker have been observed in a rat model of
T2DM (Otsuka Long-Evans Tokushima Fatty
rats) than in healthy rats (Otsuka Long-Evans
Tokushima rats), and an increase of its concentra-
tion was observed prior to the development of
hyperltration and prior to the increment of
serum creatinine concentration (Hosohata et al.
2014).
5.5 Markers of Fibrosis
and Inflammation
5.5.1 Transforming Growth Factor-b1
(TGFb1)
TGFβ1 is a cytokine and pro-brotic mediator of
kidney damage. It is secreted in an inactivated
form associated with the large latent complex
(LLC) and it has a biological effect only after it
is liberated from the LLC as active TGFβ1, a
process which takes part in the extracellular
matrix (August and Suthanthiran 2003; Lawson
et al. 2016; Sureshbabu et al. 2016; Hinz 2015).
In kidneys, increased production of active TGFβ1
leads to interstitial brosis, mesangial matrix
expansion and glomerular membrane thickening,
and, as a consequence, kidney damage and reduc-
tion in GFR (August and Suthanthiran 2003;
Sureshbabu et al. 2016; Ziyadeh 2004). Animal
models have shown that TGFβ1 could also par-
ticipate in podocyte detachment or apoptosis,
stimulating podocyte expression of VEGF that
Animal Models and Renal Biomarkers of Diabetic Nephropathy
acts in an autocrine loop, leading to increased
production of 3(IV) collagen, which probably
contributes to the thickening of the glomerular
basement membrane (Chen et al. 2004).
According to an in vitro study, TGFβ1 could
also cause oxidative stress in podocytes by itself
(Lee et al. 2003). As a consequence of podocyte
injury, proteinuria can occur (Nagata 2016).
In vitro studies have also demonstrated that
high glucose concentration stimulates TGFβ1
secretion and activation, and this marker has
been proposed as an important mediator of DN
in animal models (Ziyadeh 2004; Rocco et al.
1992; Hoffman et al. 1998). Also, in humans,
high levels of serum and urinary TGFβ1 have
been observed in a systematic review that
included T2DM patients, showing a positive cor-
relation with albuminuria (Qiao et al. 2017).
Treatment with angiotensin-converting enzyme
inhibitors has been demonstrated to decrease uri-
nary ACR and plasma TGFβ1, and the latter has
been put forward as a mechanism mediating their
nephroprotective effects (Andrésdóttir et al.
2014). Moreover, in the db/db mouse, treatment
with a neutralizing anti-TGFβ1 antibody avoids
the progression of diabetic renal hypertrophy,
mesangial matrix expansion, and the develop-
ment of renal insufciency, albeit in the absence
of a signicant reduction in albuminuria (Ziyadeh
et al. 2000).
In companion animals, this marker has been
studied to investigate DN in cats, where an incre-
ment of urinary activated TGFβ1:creatinine ratio
precedes the onset of azotemia by 6 months
(Lawson et al. 2016).
5.5.2 Vascular Endothelial Growth
Factor (VEGF)
VEGF, also named vasopermeability factor, is a
homodimeric glycoprotein with different
isoforms and heparin-binding properties. VEGF
promotes permeability, has mitogenic functions
in endothelial cells and is an important angio-
genic factor (Khamaisi et al. 2003; Neufeld et al.
1999). In the kidney, reduction of oxygen deliv-
ery contributes to inammation pathways and is
the main stimulus for VEGF expression. (Mayer
2011; Ramakrishnan et al. 2014).
In regard to DN, although in vitro studies have
demonstrated that chronic hyperglycemia is able
to increase the production of the VEGF protein
and VEGF mRNA expression (Cha et al. 2000;
Williams et al. 1997), in humans with DM it is
uncertain whether VEGF levels increase or
decrease with DN. Down-regulation of VEGF-A
mRNA expression has been observed in renal
biopsies of patients with DM, and its lower
expression was associated with podocyte loss
(Baelde et al. 2007). However, two other studies
have shown higher concentrations of plasma and
urinary VEGF in people with T1DM and T2DM,
respectively (Hovind et al. 2000; Kim et al.
2004), and higher urinary VEGF was found in
those with advanced DN. Indeed, intervention
studies also show conicting results: both VEGF
inhibition and VEGF administration improve kid-
ney function (Schrijvers et al. 2005; Kang et al.
2001).
Additionally, there is scarce information on
the role of VEGF in kidney function in compan-
ion animals, in which the study of VEGF has been
focused on its role in tumor angiogenesis
(Millanta et al. 2002; Clifford et al. 2001; Platt
et al. 2006). One study detected lower
concentrations of urinary VEGF-A creatinine
ratio in cats with CKD compared to healthy cats
(Habenicht et al. 2013).
5.5.3 Soluble Urokinase Type
Plasminogen Activator Receptor
(suPAR)
Soluble urokinase-type plasminogen activator
receptor (suPAR) is the circulating form of mem-
brane protein urokinase receptor (uPAR), which
is a glycosyl-phosphatidylinositolanchored
three-domain membrane protein that is expressed
on podocytes and other cells (immunologically
active cells and endothelial cells). Both suPAR
and uPAR regulate cell adhesion and migration
(Salim et al. 2016). Increased levels of plasma or
serum suPAR are considered independent risk
factors of cardiovascular diseases and CKD.
suPAR seems to be a reliable marker of early
L. Pérez-López et al.
CKD since its concentration increases before a
decline in GFR is identied (Salim et al. 2016). A
cohort study showed that, in patients with T1DM,
suPAR predicts cardiovascular events and a
decline in GFR, although it was not correlated
with albuminuria (Curovic et al. 2019). In con-
trast, a cross-sectional study showed a positive
correlation between suPAR concentration and
albuminuria in patients with T1DM (Theilade
et al. 2015). Likewise, in patients with T2DM,
suPAR concentration showed a positive correla-
tion with albuminuria. Indeed, it was associated
with an increased risk of new-onset
microalbuminuria in subjects at risk for T2DM
(Guthoff et al. 2017). In transgenic mice models,
it has also been shown that increased uPAR activ-
ity in podocytes leads to proteinuria (Wei et al.
2008).
5.6 Panels of Candidate Biomarkers,
Proteomic and Metabolomic
Approaches
Omicsapproaches have been used to search for
novel biomarkers and different aspects of the
pathophysiology of kidney damage (Colhoun
and Marcovecchio 2018; Carlsson et al. 2017;
Abbiss et al. 2019; Darshi et al. 2016). Each
individual biomarker of kidney disease represents
a specic pathway. Depending on these
pathways, some renal markers can be correlated
with others. Therefore, the selection of panels of
biomarkers that have low correlation with each
other could potentially be benecial for the pre-
diction of impaired kidney function (Colhoun and
Marcovecchio 2018). For example, a set of
297 biomarkers was evaluated in a recent pro-
spective study in two different cohorts of patients
with T1DM, and only two biomarkers, CD27, a
member of the tumor necrosis factor receptor
superfamily, and KIM-1 were considered to give
predictive information of DN. This yielded simi-
lar predictive power than using historical
estimated GFR and albuminuria (Colombo et al.
2019a). Similarly, another study examined
42 biomarkers in 840 serum samples of patients
with T2DM and found that prediction of the
decline in renal function could be improved by
the use of two single markers: KIM-1 and
β2 microglobulin (Colombo et al. 2019b). In con-
trast, in one study in patients with T1DM, KIM-1
did not seem to predict progression of CKD inde-
pendently of albuminuria (Panduru et al. 2015)
(Table 3).
6 Conclusions
DN is a common complication in patients with
DM and research in animal models can be useful
to fully understand the mechanisms underlying its
development and progression. However, since
various renal markers are not equally useful in
all species, further studies are required in humans.
The differences and similarities between people
and animals with DN could also bring the oppor-
tunity to investigate new treatments for DN in
humans. Although rodent models are the most
studied, other animals, such as dogs and cats,
could provide important information, since they
share the human environment.
Regarding the potential use of markers of early
kidney damage, cystatin C in humans, and
SDMA in companion animals, have already
been incorporated to the guidelines for the Evalu-
ation and Management of CKD and of the Inter-
national Renal Interest Society, respectively.
Nonetheless, many other markers have been pro-
posed, but results are conicting for most of them.
In fact, those biomarkers that have been identied
as predictors of CKD or its progression, do not
seem to add to established diagnostic tools. It is
also important to highlight that most studies
performed to date are cross-sectional in their
design. More prospective and intervention studies
are needed to replicate reported ndings and to
assess the predictive value of novel biomarkers of
DN. Indeed, large consortia such as the SysKid
(Systems Biology Towards Novel Chronic
Animal Models and Renal Biomarkers of Diabetic Nephropathy
Table 3 Epidemiological studies that provide information for the diagnosis of kidney disease or DN in both humans and animal models (histological, immunochemistry or
intervention studies were not included)
Marker Species
Author
(reference) N Study design Main results Potential clinical application
Plasma SDMA and cystatin C Humans El-khoury
et al. (2016)
40 patients who had clinical
indication for measuring GFR
Cross-
sectional
SDMA and cystatin C are
highly and inversely correlated
with GFR, more than
creatinine
SDMA and cystatin C could
detect an earlier decline in
GFR
Plasma SDMA and ADMA Humans Zobel et al.
(2017)
200 patients with T2DM Longitudinal Higher SDMA was associated
with incident cardiovascular
disease, and deterioration in
renal function
SDMA could be a marker of
DN and cardiovascular
disease in patients with
T2DM
Serum SDMA Dogs Hall et al.
(2016)
19 dogs with CKD and
20 control dogs
Retrospective SDMA detected CKD earlier
than creatinine
SDMA should also be used to
evaluate kidney function in
dogs
Serum SDMA Cats Hall et al.
(2014)
21 cats with CKD and
21 healthy control cats
Retrospective SDMA detects CKD earlier
than creatinine
SDMA should also be used to
evaluate kidney function in
cats
Serum SDMA Cats Langhorn
et al. (2018)
17 with CKD, 40 with HCM,
17 with DM, and 20 healthy
controls
Cross-
sectional
Cats with DM had
signicantly lower SDMA
concentrations than controls
SDMA is probabli not a
useful marker of DN in cats;
and further research about DN
is still needed in cats
Serum cystatin C BALB/c
mice
Song et al.
(2009)
23 partial nephrectomy
6 ischaemia reperfusion injury
model
Cross-
sectional
Cystatin C increases before
creatinine in mice with kidney
damage. CysC levels show an
earlier and sharper increase
than creatinine after bilateral
nephrectomy
Cystatin C could be a more
precise marker compared to
creatinine
8 controls
Urinary cystatin C Humans Jeon et al.
(2011)
335 T2DM patients with
normoalbuminuria (n ¼210),
those with microalbuminuria
(n ¼83) and those with
macroalbuminuria (n ¼42)
Retrospective Cystatin C was independently
associated with GFR, and was
increased in people with
diabetes, normoalbuminuria
and decreased GFR
Might predict early stages of
DN in T2DM patients
Serum and urinary Cystatin C Cats Ghys et al.
(2016)
49 cats with CKD and
41 healthy cats
Cross-
sectional
Sensitivity and specicity to
detect CKD were 22 and 100%
for Cystatin C and 83 and 93%
for creatinine
Cystatin C should not be used
to evaluated kidney function
in cats
L. Pérez-López et al.
Serum SDMA and Cystatin C Dogs Pelander
et al. (2019)
30 healthy dogs and 67 dogs
with diagnosis or suspicion of
CKD
Cross-
sectional
Creatinine and SDMA were
similar to detect reduced GFR,
whereas cystatin C was
inferior
SDMA should be measured
together with creatinine as it
might add information of
kidney function in dogs
Urinary Cystatin C- creatinine
ratio and nonalbumin protein
creatinine ratio
Humans Kim et al.
(2013)
237 T2DM patients Longitudinal After adjusting for several
clinical factors, both urinary
Cystatin C could be a useful
marker of renal function
decline in patients with
T2DM
Cystatin C- creatinine ratio
and non albuminuric protein
creatinine ratio had signicant
associations with the decline
of the estimated glomerular
ltration rate (eGFR)
β2-microglobulin, calbindin,
clusterin, EGF, GST-α,
GST-μ, KIM-1, NGAL,
osteopontin, TIMP-1, and
VEGF
Zucker
diabetic
fatty
rats
Togashi and
Miyamoto
(2013)
5 Male Zucker diabetic fatty
rats (ZDF/CrlCrlj-Leptfa/fa)
and 5 Male nondiabetic lean
rats (ZDF/CrlCrlj-Lept?/+
Cross-
sectional
Urinary levels of cystatin C,
β2-microglobulin, clusterin,
GST-μ, KIM-1 were increased
before the development of
histophatological changes
consistent with DN
Cystatin C, β2-microglobulin,
clusterin, GST-μ, KIM-1
could be used as markers of
DN in mice models of DN
Urinary RBP4 Humans Domingos
et al. (2016)
454 participants with stages
3 and 4 CKD
Cross-
sectional
A logistic regression model
showed an inverse association
between CKD-EPI eGFR and
urinary retinol binding protein
Urinary retinol binding
protein might be a promising
marker of chronic kidney
disease progression
Urinary RBP4 Humans Park et al.
(2014)
471 type 2 diabetes patients,
143 with impaired glucose
tolerance and 75 controls
Cross-
sectional
Urinary RBP4 concentration
was higher in insulin resistant
patients, and it was highly
associated with
microalbuminuria (odds ratio
2.6, 95% CI 1.64.2),
Kidney function of diabetic
patients with higher levels of
urinary RBP4 should be
evaluated closely, and it could
be useful in the management
and stratications of insulin
resistant patients. Further
investigation is needed
Serum RBP4, CysC, b2M Humans Donadio
et al. (2001)
110 patients with various
kidney diseases
Cross-
sectional
Serum concentrations of
CysC, b2M and RBP4
increase with the reduction of
GFR
Cys, b2M and RBP4 do not
seems more reliable markers
to detect a decline in kidney
function compared to
creatinine
ROC analysis showed that
diagnostic accuracy of CysC
and b2M was similar to
(continued)
Animal Models and Renal Biomarkers of Diabetic Nephropathy
Table 3 (continued)
Marker Species
Author
(reference) N Study design Main results Potential clinical application
creatinine and better than
RBP4
Plasma RBP4 Humans Cabré et al.
(2007)
165 T2DM patients Cross-
sectional
Patients with moderate renal
dysfunction (MDRD-GFR
<60 mL min
1
1.73 m
2
had
higher plasma RBP4 than
those with normal renal
function
RBP4 might predict early
decline in GFR in patients
with DN
Albuminuria was not
associated with RBP4
Serum RBP4 Humans Akbay et al.
(2010)
53 T2DM patients and
30 controls
Cross-
sectional
Logistic regression analysis
showed that microalbuminuria
is associated with increased
serum RBP4 concentration
RBP4 might predict early DN
Serum RBP4 Cats van Hoek
et al. (2008)
10 cats with CKD, 10 cats with
hyperthyroidism and
10 healthy cats
Cross-
sectional
Cats with CKD and
hyperthyroidism had higher
concentration of RBP4 than
healthy cats
RBP4 should be investigated
as a marker of impaired
kidney function in cats
Urinary and serum uromodulin Humans Prajczer
et al. (2010)
77 patients with CKD and
14 healthy subjects
Cross-
sectional
Urinary uromodulin was
positively correlated with GFR
and negatively correlated with
serum creatinine. Patients with
lower uromodulin values
showed higher degree of
tubular atrophy (assessed
through biopsy)
Urinary uromodulin might be
a early marker of tubular
damage
Serum uromodulin Humans Wiromrat
et al. (2019)
179 T1DM adolescents
patients and 61 control
subjects
Cross-
sectional
Lower levels of serum
uromodulin are associated
with albumin excretion
Serum uromodulin should be
assessed as a marker of DN
Urinary uromodulin Humans Möllsten
and Torffvit
(2010)
301 patients with T1DM,
164 with normoalbuminuria,
91 with microalbuminuria and
46 with macroalbuminuria
Cross-
sectional
Patients with albuminuria had
lower uromodulin
concentrations
Urinary uromodulin might
reect tubular damage in
patients with T1DM
L. Pérez-López et al.
Serum uromodulin Humans Fedak and
Kuźniewski
(2016)
170 patients with CKD and
30 healthy subjects
Cross-
sectional
Serum uromodulin was
inversely correlated with other
renal markers and positively
correlated with estimated GFR
Serum uromodulin might be
assessed as an early marker of
CKD
Urinary albumin excretion
ratio (AER), N-acetyl-β-D-
glucosaminidase, and the
advanced glycosylation
end-products (AGEs)
pentosidine and
AGE-uorescence
Humans Kem et al.
(2010)
55 T1DM patietns with and
110 without macroalbuminuria
91 T1DM patients with and
178 without microalbuminuria
Retrospective
case-control
N-acetyl-β-D-glucosaminidase
independently is associated
with both macroalbuminuria
and microalbuminuria. Other
markers did not independently
predict macro or
microalbuminuria.
Tubular damage occurs in
patients with T1DM and
measurement of urinary
albumin excretion ratio and
urinary N-acetyl-β-D-
glucosaminidase might
predict early impaired kidney
function
Plasma, urinary NGAL, and
urinary NGAL-to-creatinine
ratio (UNCR)
Cats Wang et al.
(2017)
80 cats with CKD and
18 healthy cats
Longitudinal NGAL values were
statistically different between
healthy cats and cats with
stage 3 or 4 CKD, however, no
statistical differents were
found between healthy cats
and cats with those with stage
2 CKD
Plasma NGAL cannot
distinguish CKD in cats
Urinary NGAL does not
detect early stages of CKD in
cats
Plasma NGAL and UNCR Dogs Steinbach
et al. (2014)
17 dogs with CKD 48 dogs
with AKI and 18 controls
subjects
Cross-
sectional
Plasma NGAL concentration
and UNCR was signicantly
higher in dogs with AKI or
CKD compared to healthy
dogs. In addition, these
markers were higher in dogs
with AKI compared with dogs
with CKD
Although NGAL is an
established marker for AKI, it
might also be useful to
distinguish dogs with CKD
from healthy dogs, although
further research is needed
Urinary NAGL, NAG and
KIM-1
Human Fu et al.
(2012b)
101 T2DM patients Cross-
sectional
All marker showed higher
levels in patients with
DM. NGAL and NAG were
positively correlated with
albuminuria. NGAL showed
signicant differences between
micro and macroalbuminuric
patients
NAGL and KIM-1 could be
early markers of DN. Those
patients with glomerular
hyperltration and higher
levels of urinary NAGL or
KIM-1, should be closely
monitored
28 control subjects
KIM-1 was not associated
with albuminuria
(continued)
Animal Models and Renal Biomarkers of Diabetic Nephropathy
Table 3 (continued)
Marker Species
Author
(reference) N Study design Main results Potential clinical application
Urinary KIM-1 and NGAL Humans de Carvalho
et al. (2016)
117 T2DM patients Cross-
sectional
Both markers were observed in
patients with T2DM with
normal or mild albuminuria,
and they were independently
associated with albuminuria
Tubular markers could help in
the early detection of DN
Urinary KIM-1, NGAL and
vanin-1
Rats Hosohata
et al. (2014)
8 male spontaneous type
2 diabetic OLETF rats and
8 male non-diabetic Long-
Evans
Cross-
sectional
Urinary KIM-1 was more
sensitive than albumin to
detect DN
KIM-1 detect early tubular
damage
Tokushima Otsuka (LETO)
rats
Urinary KIM-1 Humans Panduru
et al. (2015)
1573 T1DM patients Longitudinal
multicenter
study
Mendelian randomization
(MR) approach suggested a
causal link between increased
urinary KIM-1 and decreased
GFR. KIM-1 did not predict
progression of albuminuria
Further studies to evaluate
KIM-1 as an early marker of
DN could be interesting
Urinary active TGFβ1:
creatinine ratio
Cats Lawson
et al. (2016)
6 non-azotaemic cats that
developed azotaemia within
24 months; 6 cats and with
renal azotaemia at baseline;
and 6 non-azotemic cats
Longitudinal Increased active TGFβ1:
creatinine ratio was observed
6 month before the
development of azotemia
Urinary active TGFβ1:
creatinine ratio might be an
early marker of CKD in cats,
this marker should be
assessed in a larger sample.
Serum and urinary TGFβ1 Humans Qiao et al.
(2017)
63 case-control studies
(364 T2DM patients, 1604
T2DM and DN patients, and
2100 healthy controls)
Systematic
review
TGFβ1 levels were higher in
T2DM patients and were
positively correlated with
albuminuria
TGFβ1 could be a promising
marker of DN, although
future research is needed
Plasma uromodulin and
cystatin C
Humans Steubl et al.
(2016)
426 individuals of whom
71 healthy subjects and
355 had CKD (stages I-V)
Cross-
sectional
Multiple linear regression
modeling showed signicant
association between
uromodulin and eGFR
(coefcient estimate b.0.696,
95% condence interval
[CI] 0.6030.719, P < 0.001)
Uromodulin could be an
earlier marker of CKD
compared to creatinine and
CysC
Uromodulin was able to
differentiate between patients
in stage 0 and I
L. Pérez-López et al.
Neither cystatin C nor
creatinine distinguished
between stages 0 and I
Urinary Ig G, KIM-1, NAGL,
NAG
Humans Nauta et al.
(2011)
94 T1DM and T2DM, and
45 control subjects
Cross-
sectional
Glomerular and tubular
markers were associated with
albuminuria independently of
GFR (except KIM-1)
Markers of glomerular and
tubular damage should be
evaluated in patients with DN
Urinary RBP4,b2-
microglobulin, NAGL, NAG,
and IgG creatinine ratios
Dogs Nabity et al.
(2015)
2025 dogs with X-linked
hereditary nephropathy and
1019 dogs control subjects
Retrospective Urinary RBP4 was the most
strongly correlated with serum
creatinine and GFR. Although
logistic regression analysis
showed serum creatinine,
uIgG/c, and uB2M, but not
uRBP4/c, as signicant
independent predictors of GFR
b2-microglobulin, NAGL/c,
NAG/c, IgG/ could allow
early detection of CKD, and
RBP4/c is a marker of CKD
progression in dogs
Urinary albumin vitamin
D-binding protein, RBP4,
uromodulin,
Dogs Chakar et al.
(2017)
40 dogs with CKD and
9 control subjects
Cross-
sectional
Increased vitamin D-binding
protein and RBP4 were
detected in early stages of
CKD with or without
albuminuria
Vitamin D binding protein
and RBP4 should be studied
as an early marker of CKD,
whereas uromodulin could be
a marker of progression of
CKD in dogs
Dogs with CKD had
undetectable or lower RBP4
than controls, although
among dogs in early stages of
CKD there were no
differences
Plasma and urinary VEGF Humans Kim et al.
(2004)
147 patients with T2DM and
47 healthy controls
Cross-
sectional
VEGF concentration was
higher in T2DM patients and
was associated with
albuminuria
VEGF might be a useful
marker of DN in patients with
T2DM
Plasma VEGF Humans Hovind
et al. (2000)
199 patients with T1DM and
DN, and 188 patients with
T1DM and normoalbuminuria
Cross-
sectional
Men with DN had higher
concentration of VEGF than
normoalbuminuric patients
Sex differences might affect
VEGF concentration
Urinary cytokine (IL-8,
MCP-1. TGF- β1, VEGF):
urine creatinine ratios
Cats Habenicht
et al. (2013)
26 cats with CKD and
18 healthy cats
Cross-
sectional
Cats with CKD had a
signicantly lower urinary
levels of VEGF and higher
urinary levels of IL-8 and
Further research is needed to
evaluate the utility of measure
urinary cytokines in cats with
CKD, but it markers might
(continued)
Animal Models and Renal Biomarkers of Diabetic Nephropathy
Table 3 (continued)
Marker Species
Author
(reference) N Study design Main results Potential clinical application
TGF-β1 compared to healthy
cats
reect kidney inammaniton
and brosis
Plasma suPar Humans Salim et al.
(2016)
2292 patients from Emory
cardiovascular biobank whose
renal function was sequentially
evaluated
Longitudinal suPAR concentration
increased before a decline in
estimated GFR was observed
Kidney function of patients
with elevated suPAR should
be monitored closely
Plasma suPar Humans Curovic
et al. (2019)
667 patients with T1DM and
different levels of albuminuria
Longitudinal suPAR predicted
cardiovascular events and a
decline in GFR but was not
associated with albuminuria
suPAR is useful to detect
early DN
Plasma suPAR Humans Theilade
et al. (2015)
667 patients with T1DM and
51 control subjects
Cross-
sectional
suPAR levels were higher in
patients with cardiovascular
disease and in patients with
albuminuria. Multivariate
logistic regression analysis
showed an association
between suPAR and
albuminuria in patients with
DM
suPAR and its relations with
albuminuria should be
investigated
Plasma suPAR Humans Guthoff
et al. (2017)
258 patients at risk of T2DM Longitudinal Higher suPAR levels are
associated with an increased
risk of new-onset
microalbuminuria in subjects
at risk for type 2 diabetes
suPAR might be a useful
marker of early DN in T2DM
patients
Panel of 42 serum biomarkers Humans Colombo
et al.
(2019b)
840 patients with T2DM Longitudinal
multicenter
study
Kim-1 and β2-microglobulin
improve prediction of renal
function decline
Until further validation Kim-1
and β2-microglobulin could
be useful in clinical trials to
select those patients with
higher risk of DN
Panel of 297 serum biomarkers Humans Colombo
et al.
(2019a)
1174 patients with T1DM Longitudinal
multicenter
study
Predictive information can be
obtained using just two
biomarkers (CD27 and
KIM-1)
Few biomarkers are necessary
to gain prediction of DN
diagnosis.
Panel of 207 serum biomarkers Humans Looker et al.
(2015)
154 cases (40% reduction in
GFR) and 153 controls
Longitudinal
multicenter
study
14 biomarkers were associated
with CKD progression
(SDMA, SDMA/ADMA ratio,
These panel detected some
novel biomarkers that require
further investigation
L. Pérez-López et al.
Kim-1, creatinine, β2-
Microglobulin, α1 Antitrypsin,
Uracil, N-terminal
prohormone of brain
natriuretic peptide,
C16-acylcarnitine,
Hydroxyproline, Fibroblast
growth factor-21, Fatty acid-
binding protein heart,
Creatine, Adrenomedullin)
Panel of 402 plasma
biomarkers
Humans Kammer
et al. (2019)
481 T2DM patients with
incident or early CKD
(comparing patients with
stable GFR and patients with a
rapid decline of GFR)
Longitudinal
multicenter
study
KIM-1 was the most important
predictor of GFR decline
KIM-1 might be a useful
marker of DN in patients with
T2DM
Abbreviations: eGFR estimated glomerular ltration rate, DN diabetic nephropathy, T1DM patients with type 1 diabetes, T2DM patients with type 2 diabetes, SDMA symmetric
dimethylarginine, NGAl neutrophil gelatinase-associated lipocalin, AER Albumin excretion ratio, AGEs advanced glycosylation end-products, AKI acute kidney injury, CKD
chronic kidney disease, DN diabetic nephropathy, UNCR urinary NGAL-to-creatinine ratio, KIM-1, kidney injury molecule 1, IgG immunoglobulin G, MCP-1 urinary
monocyte chemoattractant protein-1, NAG N-acetyl-β-D-glucosaminidase, GST-αalpha glutation S transferasa, GST-μmu glutation S transferasa, IL-8 interleukin 8, TIMP-1
tissue inhibitor of metalloprotease-1, VEGF vascular endothelial growth factor, RBP4 retinol binding protein 4, suPAR soluble urokinase type plasminogen activator receptor,
TGFβ1transforming growth factor β-1
Animal Models and Renal Biomarkers of Diabetic Nephropathy
Kidney Disease Diagnosis and Treatment), SUM-
MIT (Surrogate markers for micro and
macrovascular hard endpoints for innovative dia-
betes tools) and BEAt-DKD (Biomarker Enter-
prise to Attack Diabetic Kidney Disease) are
providing and will provide important results in
the near future.
References
Abbiss H, Maker GL, Trengove RD (2019) Metabolomics
approaches for the diagnosis and understanding of
kidney diseases. Meta 9(2):E34
Akbay E, Muslu N, Nayir E, Ozhan O, Kiykim A (2010)
Serum retinol binding protein 4 level is related with
renal functions in type 2 diabetes. J Endocrinol
Investig 33(10):725729
Al-Ghazlat SA, Cathy B, Langston E, Greco DS, Reine
NJ, May SN et al (2011) The prevalence of
microalbuminuria and proteinuria in cats with diabetes
mellitus. Top Companion Anim Med 26(3):154157
Almy FS, Christopher MM, King DP, Brown SA (2002)
Evaluation of cystatin C as an endogenous marker of
glomerular ltration rate in dogs. J Vet Intern Med 16
(1):4551
Alpers CE, Hudkins KL (2011) Mouse models of diabetic
nephropathy. Curr Opin Nephrol Hypertens 20
(3):278284
American Diabetes Association (2004) Nephropathy in
diabetes. Diabetes Care 27(Suppl 1):S79S83
Andrésdóttir G, Jensen ML, Carstensen B, Parving HH,
Rossing K, Hansen TW et al (2014) Improved survival
and renal prognosis of patients with type 2 diabetes and
nephropathy with improved control of risk factors.
Diabetes Care 37(6):16601667
Antic V, Tempini A, Montani JP (1999) Serial changes in
cardiovascular and renal function of rabbits ingesting a
high-fat, high-calorie diet. Am J Hypertens 12(8 Pt
1):826829
August P, Suthanthiran M (2003) Transforming growth
factor beta and progression of renal disease. Kidney Int
Suppl 87:S99S104
Baelde HJ, Eikmans M, Lappin DW, Doran PP,
Hohenadel D, Brinkkoetter PT et al (2007) Reduction
of VEGF-A and CTFG expression in diabetic nephrop-
athy is associated with podocyte loss. Kidney Int 71
(7):637645
Barlett PC, Van Buren JW, Barlett AD, Zhou C (2010)
Case-control study of risk factors associated with
canine and feline chronic kidney disease. Vet Med Int
2010:957570
Basi S, Fesler P, Mimran A, Lewis LB (2008)
Microalbuminuria in type 2 diabetes and hypertension.
Diabetes care 2008. Diabetes Care 31(Suppl 2):S194
S201
Bates JM, RafHM, Prasadan K, Mascarenhas R,
Laszik Z, Maeda N (2004) Tamm-Horsfall protein
knockout mice are more prone to urinary tract infec-
tion: rapid communication. Kidney Int 65:791797
Betz B, Conway BR (2014) Recent advances in animal
models of diabetic nephropathy. Nephron Exp Nephrol
126:191195
Blantz RC (1998) Pathophysiology of pre-renal azotemia.
Kidney Int 53:512523
Bolignano D, Donato V, Lacquaniti A, Fazio MR, Bono C,
Coppolino G et al (2010) Neutrophil gelatinase-
associated lipocalin (NGAL) in human neoplasias: a
new protein enters the scene. Cancer Lett 288:1016
Brito-Casillas Y, Melián C, Wägner AM (2016) Study of
the pathogenesis and treatment of diabetes mellitus
through animal models. Endocrinol Nutr 63
(7):345353
Cabré A, Lázaro I, Girona J, Manzanares J, Marimón F,
Plana N et al (2007) Retinol-binding protein 4 as a
plasma biomarker of renal dysfunction and cardiovas-
cular disease in type 2 diabetes. J Intern Med 262
(4):496503
Câmara NO, Iseki K, Kramer H, Liu ZH, Sharma K (2017)
Kidney disease and obesity: epidemiology,
mechanisms and treatment. Nat Rev Nephrol 13
(3):181190
Can A, Bekpinar S, Gurdol F, Tutuncu Y, Unlucerci Y,
Dinccag N (2011) Dimethylarginines in patients with
type 2 diabetes mellitus: relation with the glycaemic
control. Diabetes Res Clin Pract 94(3):e61e64
Caramori ML, Fioretto P, Mauer M (2003) Low glomeru-
lar ltration rate in normoalbuminuric type 1 diabetic
patients: an indicator of more advanced glomerular
lesions. Diabetes 52(4):10361040
Carlsson AC, Ingelsson E, Sundström J, Carrero JJ,
Gustafsson S, Feldreich T et al (2017) Use of proteo-
mics to investigate kidney function decline over
5 years. Clin J Am Soc Nephrol 12(8):12261235
Castrop H, Schießl IM (2014) Physiology and pathophys-
iology of the renal Na-K-2Cl cotransporter (NKCC2).
Am J Physiol Renal Physiol 307(9):F991F1002
Cha DR, Kim NH, Yoon JW, Jo SK, Cho WY, Kim HK
et al (2000) Role of vascular endothelial growth factor
in diabetic nephropathy. Kidney Int Suppl 77:S104
S1012
Chakar F, Kogika M, Sanches TR (2017) Urinary Tamm-
Horsfall protein, albumin, vitamin D-binding protein,
and retinol-binding protein as early biomarkers of
chronic kidney disease in dogs. Phys Rep 5(11):
e13262
Chakraborty J, Below AA, Solaiman D (2004) Tamm-
Horsfall protein in patients with kidney damage and
diabetes. Urol Res 32(2):7983
Chandler M, Cunningham S, Lund EM, Khanna C,
Naramore R et al (2017) Obesity and associated
comorbidities in people and companion animals- a
one health perspective. J Comp Pathol 156(4):296309
Chen S, Kasarma Y, Lee JS, Jim B, Marin M, Ziyadeh FN
(2004) Podocyte-derived vascular endothelial growth
L. Pérez-López et al.
factor mediates the stimulation of 3(IV) collagen pro-
duction by transforming growth factor-1 in mouse
podocytes. Diabetes 52(11):29392949
Cheuiche AV, Queiroz M, Azeredo-da-Silva ALF,
Silveiro SP (2019) Performance of cystatin C-based
equations for estimation of glomerular ltration rate
in diabetes patients: a Prisma-compliant systematic
review and Meta-analysis. Sci Rep 9:1418
Christensen EI, Moskaug JO, Vorum H (1999) Evidence
for an essential role of megalin in transepithelial trans-
port of retinol. J Am Soc Nephrol 10(4):685695
Clifford CA, Hughes D, Beal MW, Mackin AJ, Henry CJ,
Shofer FS et al (2001) Plasma vascular endothelial
growth factor concentrations in healthy dogs and
dogs with hemangiosarcoma. J Vet Intern Med 15
(2):131135
Closs EI, Basha FZ, Habermeier A, Förstermann U (1997)
Interference of L-arginine analogues with L-arginine
transport mediated by the y/carrier hCAT-2B. Nitric
Oxide 1(1):6573
Colhoun HM, Marcovecchio ML (2018) Biomarkers of
diabetic kidney disease. Diabetologia 61(5):9961011
Colombo M, Valo E, McGurnaghan SJ, Sandholm N,
Blackbourn LAK, Dalton RN et al (2019a) Biomarker
panels associated with progression of renal disease in
type 1 diabetes. Diabetologia 62(9):16161627
Colombo M, Looker HC, Farran B, Hess S, Groop L,
Palmer CAN et al (2019b) Serum kidney injury mole-
cule 1 and β
2
-microglobulin perform as well as larger
biomarker panels for prediction of rapid decline in
renal function in type 2 diabetes. Diabetologia 62
(1):156158
Curovic VR, Theilade S, Winther SA, Tofte N, Eugen-
Olsen J, Persson F et al (2019) Soluble Urokinase
plasminogen activator receptor predicts cardiovascular
events, kidney function decline, and mortality in
patients with type 1 diabetes. Diabetes Care 42
(6):11121119
DAmico G, Bazzi C (2003) Pathophysiology of protein-
uria. Kidney Int 63(3):809825
Darshi M, Van Espen B, Sharma K (2016) Metabolomics
in diabetic kidney disease: unraveling the biochemistry
of a silent killer. Am J Nephrol 44(2):92103
De Carvalho JAM, Tatsch E, Hausen BS, Bollick Y,
Moretto MB, Duarte T et al (2016) Urinary kidney
injury molecule-1 and neutrophil gelatinase-associated
lipocalin as indicators of tubular damage in
normoalbuminuric patients with type 2 diabetes. Clin
Biochem 49(3):232236
De Francesco PN, Cornejo MP, Barrile F, García
Romero G, Valdivia S, Andreoli MF et al (2019)
Inter-individual variability for high fat diet consump-
tion in inbred C57BL/6 mice. Front Nutr 6:67
Delanaye P, Cavalier E, Pottel H (2017) Serum creatinine-
not so simple! Nephron 136(4):302308
Diabetes Complications Consortium (DiaComp) (2003)
Validation of mouse models of diabetic nephropathy
[Internet]. EEUU: Diabetes Complications Consortium
(DiaComp); [Cited 2019 July 24]. Available from:
https://www.diacomp.org/shared/document.aspx?
id¼25&docType¼Protocol
Domingos MA, Moreira SR, Gomez L, Goulart A, Lotufo
PA, Benseñor I et al (2016) Urinary retinol-binding
protein: relationship to renal function and cardiovascu-
lar risk factors in chronic kidney disease. PLoS One 11
(9):e0162782
Donadio C, Lucchesi A, Ardini M, Giordani R (2001)
Cystatin C, b2-microglobulin, and retinol-binding pro-
tein as indicators of glomerular ltration rate- compar-
ison with plasma creatinine. J Pharm Biomed Anal 24
(56):835842
Dronavalli S, Duka I, Bakris GL (2008) The pathogenesis
of diabetic nephropathy. Nat Clin Pract Endocrinol
Metab 4(8):444452
Dwyer TM, Banks SA, Alonso-Galicia M (2000) Distri-
bution of renal medullary hyaluronan in lean and obese
rabbits. Kidney Int 58(2):721729
El-khoury JM, Bunch DR, Hu B, Payto D, Reineks EZ,
Wang S (2016) Comparison of symmetric
dimethylarginine with creatinine, cystatin C and their
eGFR equations as markers of kidney function. Clin
Biochem 49(15):11401143
Engeli S, Böhnke J, Gorzelniak K, Janke J, Schiling P,
Bader M et al (2005) Weight loss and the renin-
angiotensin-aldosterone system. Hypertension 45
(3):356362
Fedak D, Kuźniewski M, Fugiel A (2016) Serum
uromodulin concentrations correlate with glomerular
ltration rate in patients with chronic kidney disease.
Pol Arch Med Wewn 126(12):9951004
Ferguson MA, Waikar SS (2012) Established and
emerging markers of kidney function. Clin Chem 58
(4):680689
Finch NC, Syme HM, Elliott J (2018) Development of an
estimated glomerular ltration rate formula in cats. J
Vet Intern Med 32(6):19701976
Fioretto P, Mauer M (2007) Histopathology of diabetic
nephropathy. Semin Nephrol 27(2):195207
Fjaertoft G, Foucard T, Xu S, Venge P (2005) Human
neutrophil lipocalin (HNL) as a diagnostic tool in chil-
dren with acute infections: a study of the kinetics. Acta
Paediatr 94:661666
Forbes JM, Cooper ME (2007) Diabetic nephropathy:
where hemodynamics meets metabolism. Exp Clin
Endocrinol Diabetes 115:6984
Fricker M, Wiesli P, Brändle M, Schwegler B, Schmind C
(2003) Impact of thyroid dysfunction on serum cystatin
C. Kidney Int 63(5):19441947
Fu WJ, Xiong SL, Fang YG, Wen S, Chen ML, Deng RT
et al (2012a) Urinary tubular biomarkers in short-term
type 2 diabetes mellitus patients: a cross-sectional
study. Endocrine 41(1):8288
Fu WJ, Li BL, Wang SB, Chen ML, Deng RT, Ye CQ et al
(2012b) Changes of the tubular markers in type 2 dia-
betes mellitus with glomerular hyperltration. Diabetes
Res Clin Pract 95(1):105109
Ghys LFE, Paepe D, Lefebvre HP, Reynolds BS,
Croubels S, Meyer E et al (2016) Evaluation of
Animal Models and Renal Biomarkers of Diabetic Nephropathy
Cystatin C for the detection of chronic kidney disease
in cats. J Vet Intern Med 30:10741082
Giacchetti G, Faloia E, Mariniello B, Sardu C, Gatti C,
Camilloni MA et al (2002) Overexpression of the
renin-angiotensin system in human visceral adipose
tissue in normal and overweight subjects. Am J
Hypertens 15(5):381382
Giasson J, Li GH, Chen Y (2011) Neutrophil gelatinase-
associated lipocalin (NGAL) as a new biomarker for
non-acute kidney injury (AKI) diseases. Inamm
Alerrgy Drug Targets 10(4):272282
Graham TE, Yang Q, Blüher M et al (2006) Retinol-
binding protein 4 and insulin resistance in lean,
obese, and diabetic subjects. N Engl J Med 354
(24):25522563
Greene JP, Lefebvre SL, Wang M, Wang M, Yang M,
Lund EM et al (2014) Risk factors associated with the
development of chronic kidney disease in cats
evaluated at primary care veterinary hospitals. J Am
Vet Med Assoc 244(3):320327
Gross JL, de Azevedo MJ, Silveiro SP, Canani LH,
Caramori ML, Zelmanovitz T (2005) Diabetic
nephropathy: diagnosis, prevention, and treatment.
Diabetes Care 28(1):164176
Guthoff M, Wagner R, Randrianarisoa E, Hatziagelaki E,
Peter A, Häring HU et al (2017) Soluble urokinase
receptor (suPAR) predicts microalbuminuria in
patients at risk for type 2 diabetes mellitus. Sci Rep
7:40627
Haase M, Haase-Fielitz A, Bellomo R, Mertens PR (2011)
Neutrophil gelatinase-associated lipocalin as a marker
of acute renal disease. Curr Opin Hematol 18:1118
Habenicht LM, Webb TL, Clauss LA, Dow SW, Quimby
JM (2013) Urinary cytokine levels in apparently
healthy cats and cats with chronic kidney disease. J
Feline Med Surg 15(2):99104
Hall JA, Yerramilli M, Obare E, Yerramilli M, Yu S,
Jewell DE (2014a) Comparison of serum
concentrations of symmetric dimethylarginine and cre-
atinine as kidney function biomarkers in healthy geri-
atric cats fed reduced protein foods enriched with sh
oil, L-carnitine, and medium-chain triglycerides. Vet J
202(3):588596
Hall JA, Yerramilli M, Obare E, Yerramilli M, Jewell DE
(2014b) Comparison of serum concentrations of sym-
metric dimethylarginine and creatinine as kidney func-
tion biomarkers in cats with chronic kidney disease. J
Vet Intern Med 28(6):16761683
Hall JA, Yerramilli M, Obare E, Yerramilli M, Melendez
LD, Jewell DE (2015) Relationship between lean body
mass and serum renal biomarkers in healthy dog. J Vet
Intern Med 29(3):808814
Hall JA, Yerramilli M, Obare E, Yerramilli M, Almes K,
Jewell DE (2016) Serum concentrations of symmetric
dimethylarginine and creatinine in dogs with naturally
occurring chronic kidney disease. J Vet Intern Med 30
(3):794802
Hans-Joachim A, Davis JM, Thurau K (2016) Nephron
protection in diabetic kidney disease. N Engl J Med
375:20962098
Henegar JR, Bigler SA, Henegar LK, Tyagi SC, Hall JE
(2001) Functional and structural changes in the kidney
in the early stages of obesity. J Am Soc Nephrol 12
(6):12111217
Henze A, Frey SK, Raila J, Tepel M, Scholze A, Pfeiffer
AF et al (2008) Evidence that kidney function but not
type 2 diabetes determines retinol-binding protein
4 serum levels. Diabetes 57(12):33233326
Hinz B (2015) The extracellular matrix and transforming
growth factor-β1 tale of a strained relationship. Matrix
Biol 47:5465
Hoenig M (2012) The cat as a model for human obesity
and diabetes. J Diabetes Sci Technol 6(3):525533
Hoffman BB, Sharma K, Zhu Y, Ziyadeh FN (1998)
Transcriptional activation of transforming growth
factor-beta1 in mesangial cell culture by high glucose
concentration. Kidney Int 54(4):11071116
Hokamp JA, Nabity MB (2016) Renal biomarkers in
domestic species. Vet Clin Pathol 45(1):2856
Hoshi S, Shu Y, Yoshida F, Inagaki T, Sonoda J,
Watanabe T et al (2002) Podocyte injury promotes
progressive nephropathy in Zucker diabetic fatty rats.
Lab Investig 82(1):2535
Hosohata K, Ando H, Takeshita Y, Misu H, Takamura T,
Kaneko S et al (2014) Urinary Kim-1 is a sensitive
biomarker for the early stage of diabetic nephropathy
in Otsuka Long-Evans Tokushima Fatty rats. Diab
Vasc Dis Res 11(4):243250
Hovind P, Tarnow L, Oestergaard PB, Parving HH (2000)
Elevated vascular endothelial growth factor in type
1 diabetic patients with diabetic nephropathy. Kidney
Int Suppl 75:S56S61
Hudkins KL, Pichaiwong W, Wietecha T, Kowalewska J,
Banas MC, Spencer MW et al (2010) BTBR Ob/Ob
mutant mice model progressive diabetic nephropathy. J
Am Soc Nephrol 21(9):15331542
Ingvorsen C, Karp NA, Lelliott CJ (2017) The role of sex
and body weight on the metabolic effects of high-fat
diet in C57BL/6N mice. Nutr Diabetes 7(4):e261
International Renal Interest Society (IRIS) (2015a) GFR in
practice: urine specic gravity. [Internet]. International
Renal Interest Society (IRIS), Vienna; [Cited 2019 July
25]. Available from: http://www.iris-kidney.com/edu
cation/urine_specic_gravity.html
International Renal Interest Society (IRIS) (2015b) GFR in
practice: assessment of glomerular ltration rate in
dogs. [Internet]. International Renal Interest Society
(IRIS), Vienna; [Cited 2019 July 25]. Available from:
http://www.iris-kidney.com/education/gfr_in_practice.
html
International Renal Interest Society (IRIS) (2017) IRIS
staging of CKD [Internet]. International Renal Interest
Society (IRIS), Vienna; [Cited 2019 May 22]. Avail-
able from: http://www.iris-kidney.com/guidelines/
staging.html
L. Pérez-López et al.
Jeon YK, Kim MR, Huh JE, Mok JY, Song SH, Kim SS
et al (2011) Cystatin C as an early biomarker of
nephropathy in patients with type 2 diabetes. J Korean
Med Sci 26:258263
Jordan E, Kley S, Le NA, Waldron M, Hoenig M (2008)
Dyslipidemia in obese cats. Domest Anim Endocrinol
35(3):290299
Kachapati K, Adams D, Bednar K, Ridgway WM (2012)
The Non-Obese Diabetic (NOD) mouse as a model of
human type 1 diabetes. Methods Mol Biol 933:316
Kammer M, Heinzel A, Willency JA, Dufn KL, Mayer G,
Simons K et al (2019) Integrative analysis of prognos-
tic biomarkers derived from multiomics panels helps
discrimination of chronic kidney disease trajectories in
people with type 2 diabetes. Kidney Int 96
(6):13811388
Kanasaki K, Gangadhar T, Koya D (2013) Diabetic
nephropathy: the role of inammation in broblast
activation and kidney brosis. Front Endocrinol
(Lausanne) 4:7
Kang DH, Hughes J, Mazzali M, Schreiner GF, Johnson
RJ (2001) Impaired angiogenesis in the remnant kid-
ney model: II. Vascular endothelial growth factor
administration reduces renal brosis and stabilizes
renal function. J Am Soc Nephrol 12(7):14461457
Kaseda R, Iino N, Hosojima M, Takea T, Hosaka K,
Kobayashi A (2007) Megalin-mediated endocytosis
of cystatin C in proximal tubule cells. Biochem
Biophys Res Commun 357(4):11301134
Kavarikova S (2018) Indirect markers of glomerular ltra-
tion rate in dogs and cats: a review. Vet Med 63
(9):395412
Kawano K, Hirashima T, Mori S, Saitho Y, Kurosumi M,
Natori T (1992) Spontaneous long-term hyperglycemic
rat with diabetic complications. Diabetes 41
(11):14221428
Kawano K, Hirashima T, Mori S, Natori T (1994) OLETF
(Otsuka Long-Evans Tokushima Fatty) rat: a new
NIDDM rat strain. Diabetes Res Clin Pract 24
(Suppl):S317S320
Keatings VM, Barnes PJ (1997) Granulocyte activation
markers in induced sputum: comparison between
chronic obstructive pulmonary disease, asthma, and
normal subjects. Am J Respir Crit Care Med
155:449453
Kem EF, Erhard P, Sun W, Genuth S, Weiss MF (2010)
Early urinary markers of diabetic kidney disease: a
nested case-control study from the Diabetes Control
and Complications Trial (DCCT). Am J Kidney Dis 55
(5):824834
Kern PA, Ranganathan S, Li C, Li C, Wood L,
Ranganathan G (2001) Adipose tissue tumor necrosis
factor and interleukin-6 expression in human obesity
and insulin resistance. Am J Physiol Endocrinol Metab
280(5):E745E751
Khamaisi M, Schrijivers BF, De Vriese AS, Itamar R,
Flyvbjerg A et al (2003) The emerging role of VEGF
in diabetic kidney disease. Nephrol Dial Transplant 18
(8):14271430
Kim NH, Kim KB, Kim DL, Kim SG, Choi KM, Baik SH
et al (2004) Plasma and urinary vascular endothelial
growth factor and diabetic nephropathy in type 2 diabe-
tes. Diabet Med 21(6):545551
Kim SS, Song SH, Kim IJ, Jeon YK, Kim BH, Kwak IS
et al (2013) Urinary cystatin C and tubularproteinuria
predict progression of diabetic nephropathy. Diabetes
Care 36:656661
Kitada M, Ogura Y, Koya D (2016) Rodent models of
diabetic nephropathy: their utility and limitations. Int J
Nephrol Renov Dis 9:279290
Kivarikova S (2015) Urinary biomarkers of renal function
in dogs and cats: a review. Vet Med 60(11):589602
Lamacchia O, Viazzi F, Fioretto P et al (2018)
Normoalbuminuric kidney impairment in T1DM
patients. Diabetol Metab Syndr 10:60
Langhorn R, Kieler IN, Koch J, Christiansen LB, Jessen
LR (2018) Symmetric dimethylarginine in cats with
hypertrophic cardiomyopathy and diabetes mellitus. J
Vet Intern Med 32:5763
Lawson JS, Syme HM, Wheeler-Jones CP, Elliot J (2016)
Urinary active transforming growth factor βin feline
chronic kidney disease. Vet J 214:16
Lee HB, Yu MR, Yang Y, Jiang Z, Ha H (2003)
Transforming growth factor β1-induced apoptosis in
podocytes via the extracellular signal-regulated
kinase-mammalian target of rapamycin complex
1-NADPH oxidase 4 Axis. J Am Soc Nephrol 14
(8 Suppl 3):S241S245
Levey AS, Coresh J, Balk E et al (2003) National kidney
foundation practice guidelines for chronic kidney dis-
ease: evaluation, classication, and stratication. Ann
Intern Med 139(2):137147
Levin A, Steven PE, Bilous RW, Coresh J, De Francisco
ALM, De Jong PE et al (2013) Kidney disease:
improving global outcomes (KDIGO) CKD work
group. KDIGO 2012 clinical practice guideline for
the evaluation and management of chronic kidney dis-
ease. Kidney Int Suppl 3(1):1150
Li Z, Woollard JR, Wang S, Korsmo MJ, Ebrahimi B,
Grande JP et al (2011) Increased glomerular ltration
rate in early metabolic syndrome is associated with
renal adiposity and microvascular proliferation. Am J
Physiol Renal Physiol 301(5):F1078F1087
Liu Y, Mo L, Goldfarb DS, Evan AP, Liang F, Khan SR
et al (2010) Progressive renal papillary calcication
and ureteral stone formation in mice decient for
Tamm-Horsfall protein. Am J Physiol Renal Physiol
299:F469F478
Looker HC, Colombo M, Hess S, Brosnan MJ, Farran B,
Dalton RN et al (2015) Biomarkers of rapid chronic
kidney disease progression in type 2 diabetes. Kidney
Int 88(4):888896
LovrenčićMV, Biljak ZR, BožičevićS, Prašek M,
PavkovićP, Knotek M (2012) Estimating glomerular
ltration rate (GFR) in diabetes: the performance of
MDRD and CKD-EPI equations in patients with vari-
ous degrees of albuminuria. Clin Biochem 45
(18):16941696
Animal Models and Renal Biomarkers of Diabetic Nephropathy
MacIsaac RJ, Tsalamandris C, Panagiotopoulos S, Smith
TJ, McNeil KJ, Jerums G (2004) Non-albuminuric
renal insufciency in T2DM. Diabetes Care 27
(1):195200
Marcovecchio ML, Dalton RN, Turner C, Prevost AT,
Widmer B, Amin R et al (2010) Symmetric
dimethylarginine, an endogenous marker of glomerular
ltration rate, and the risk for microalbuminuria in
young people with type 1 diabetes. Arch Dis Child
95(2):119124
Marynissen SJJ, Pascale MYS, Ghys LFE, Paepe D,
Delanghe J, Galac S et al (2016) Long-term follow-
up of renal function assessing serum cystatin C in dogs
with diabetes mellitus or hyperadrenocorticism. Vet
Clin Pathol 452(2):320329
Mayer G (2011) Capillary rarefaction, hypoxia, VEGF and
angiogenesis in chronic renal disease. Nephrol Dial
Transplant 26(4):11321137
McDermott JR (1976) Studies on the catabolism of
NG-methylargire, NG, NlG-dimtylarginie and; NG,
NG dimethylarginine in the rabbit. Biochem J 154
(1):179184
Millanta F, Lazzeri G, Vannozzi I, Viacava P, Poli A
(2002) Correlation of vascular endothelial growth fac-
tor expression to overall survival in feline invasive
mammary carcinomas. Vet Pathol 39(6):690696
Mogensen CE, Christensen CK, Vittinghus E (1983) The
stages in diabetic renal disease with emphasis on the
stage of incipient diabetic nephropathy. Diabetes 32
(Suppl. 2):6478
Mohapatra J, Sharma M, Acharya A, Pandya G,
Chatterjee A, Balaraman R et al (2011) Retinol-
binding protein 4: a possible role in cardiovascular
complications. Br J Pharmacol 164(8):19391948
Möllsten A, Torffvit O (2010) Tamm-Horsfall protein
gene is associated with distal tubular dysfunction in
patients with type 1 diabetes. Scand J Urol Nephrol 44
(6):438434
Moresco RN, Bochi GV, Stein CS, De Carvalho JAM,
Cembranel BM, Bollick YS (2018) Urinary kidney
injury molecule-1 in renal disease. Clin Chim Acta
487:1521
Mussap M, Plebani M (2004) Increased biochemistry and
clinical role of human cystatin C. Crit Rev Clin Lab Sci
41(56):467550
Nabity MB, Lees GE, Cianciolo R, Boggess MM, Steiner
JM, Suchodolski JS (2012) Urinary biomarkers of
renal disease in dogs with X-linked hereditary
nephropathy. J Vet Intern Med 26(2):282293
Nabity MB, Lees GE, Boggess MM, Yerramilli M,
Obare E, Yerramilli M et al (2015) Symmetric
dimethylarginine assay validation, stability, and evalu-
ation as a marker for the early detection of chronic
kidney disease in dogs. J Vet Intern Med 29
(4):10361044
Nagata M (2016) Podocyte injury and its consequences.
Kidney Int 89(6):12211230
Nakagawa T, Sato W, Glushakova O, Heinig M, Clarke T,
Campbell-Thompson M et al (2007) Diabetic
endothelial nitric oxide synthase knockout mice
develop advanced diabetic nephropathy. J Am Soc
Nephrol 18(2):539550
Narita T, Hosoba M, Kakei M (2006) ItoS. Increased
urinary excretions of immunoglobulin G, ceruloplas-
min, and transferrin predict development of
microalbuminuria in patients with type 2. Diabetes
Care 29(1):142144
National Institute of Diabetes and Digestive and Kidney
Disaese (NIDDK) (2019) Glomerular disease primer:
the normal kidney (Kidney anatomy and physiology)
[Internet]. National Institute of Diabetes and Digestive
and Kidney Disaese (NIDDK), Phoenix; [Cited
2020 March 13]. Available from: https://www.niddk.
nih.gov/research-funding/at-niddk/labs-branches/kid
ney-diseases-branch/kidney-disease-section/glomeru
lar-disease-primer/normal-kidney
Nauta FL, Wendy EB, Bakker SJL et al (2011) Glomerular
and tubular damage markers are elevated in patients
with diabetes. Diabetes Care 34(4):975981
Nelson RW, Reusch CE (2014) Animal models of disease:
classication and etiology of diabetes in dogs and cats.
J Endocrinol 222(3):T1T9
Neufeld G, Cohen T, Gengrinovithch S, Poltorak Z (1999)
Vascular endothelial growth factor (VEGF) and its
receptors. FASEB J 13(1):922
Nielsen SE, Andersen S, Zdunek D, Andersen S,
Zdunek D, Hess G et al (2011) Tubular markers do
not predict the decline in glomerular ltration rate in
type 1 diabetic patients with overt nephropathy. Kid-
ney Int 79(10):11131118
Nijveldt RJ, Teerlink T, Siroen MP, van Lambalgen AA,
Rauwerda JA, van Leeuwen PA (2003) The liver is an
important organ in the metabolism of asymmetrical
dimethylarginine (ADMA). Clin Nutr 22(1):1722
Novellas R, Ruiz de Gopegui R, Espada Y (2010) Assess-
ment of renal vascular resistance and blood pressure in
dogs and cats with renal disease. Vet Rec 166
(20):618623
Paepe D, Ghys LF, Smets P, Lefebvre HP, Croubels S,
Daminet S (2015) Routine kidney variables, glomeru-
lar ltration rate and urinary cystatin C in cats with
diabetes mellitus, cats with chronic kidney disease and
healthy cats. J Feline Med Surg 17(10):880888
Panduru NM, Sandholm N, Forsblom C, Saraheimo M,
Dahlström EH, Thorn LM et al (2015) Kidney injury
molecule-1 and the loss of kidney function in diabetic
nephropathy: a likely causal link in patients with type
1 diabetes. Diabetes Care 38(6):11301137
Park SE, Lee NS, Park JW, Rhee EJ, Lee WY, Oh KW
et al (2014) Association of urinary RBP4 with insulin
resistance, inammation, and microalbuminuria. Eur J
Endocrinol 171(4):443449
Pavenstädt H (2000) Roles of the podocyte in glomerular
function. Am J Physiol Renal Physiol 278(2):F173
F179
Payne JR, Brodbelt DC, Fuentes VL (2017) Blood pres-
sure measurements in 780 apparently healthy cats. J
Vet Intern Med 31(1):1521
L. Pérez-López et al.
Pelander L, Häggström J, Larsson A (2019) Comparison
of the diagnostic value of symmetric dimethylarginine,
cystatin C, and creatinine for detection of decreased
glomerular ltration rate in dogs. J Vet Intern Med 33
(2):630639
Pérez-López L, Boronat M, Melián C, Saavedra P, Brito-
Casillas Y, Wägner AM (2019) Assessment of the
association between diabetes mellitus and chronic kid-
ney disease in adult cats. J Vet Intern Med 33
(5):19211925
Peti-Peterdi J, Harris RC (2010) Macula Densa sensing
and signaling mechanisms of renin release. J Am Soc
Nephrol 21(7):10931096
Platt SR, Scase TJ, Adams V, Adams V, Wieczorek L,
Miller J et al (2006) Vascular endothelial growth factor
expression in canine intracranial meningiomas and
association with patient survival. J Vet Intern Med 20
(3):663668
Potočnjak I, RadulovićB, Degoricija V, TrbušićM,
Pregarner G, Berghold A et al (2018) Serum
concentrations of asymmetric and symmetric
dimethylarginine are associated with mortality in
acute heart failure patients. Int J Cardiol 261:109113
Prajczer S, Heidenreich U, Pfaller W, Kotanko P,
Lhotta K, Jennings P (2010) Evidence for a role of
uromodulin in chronic kidney disease progression.
Nephrol Dial Transplant 25(6):19861903
Premaratne E, Verma S, Ekinci EI, Theverkalam G,
Jerums G, Maclsaac RJ (2015) The impact of
hyperltration on the diabetic kidney. Diabetes Metab
41(1):517
Priyanka M, Jeyaraja K, Thirunavakkarasu PS (2018)
Abnormal renovascular resistance in dogs with diabe-
tes mellitus: correlation with glycemic status and pro-
teinuria. Iran J Vet Res 19(4):304309
Qiao YC, Chen YL, Pan YH, Ling W, Tian F, Zhan XX
et al (2017) Changes of transforming growth factor
beta 1 in patients with type 2 diabetes and diabetic
nephropathy: a PRISMA-compliant systematic review
and meta-analysis. Medicine (Baltimore) 96(15):e6583
Ramakrishnan S, Anand V, Roy S (2014) Vascular endo-
thelial growth factor signaling in hypoxia and inam-
mation. J NeuroImmune Pharmacol 9(2):142160
Rampoldi L, Scolari F, Amoroso A, Ghiggeri G, Devuyst
O (2011) The rediscovery of uromodulin (Tamm
Horsfall protein) from tubulointerstitial nephropathy
to chronic kidney disease. Kidney Int 80(4):338347
Rayner H, Thomas M, Milford D (2016) Kidney anatomy
and physiology. In: Understanding kidney diseases, 1st
edn. Springer, Cham, pp 110
Relford R, Robertson J, Clements C (2016) Symmetric
dimethylarginine improving the diagnosis and staging
of chronic kidney disease in small animals. Vet Clin
North Am Small Anim Pract 46(6):941960
Riedel J, Badewien-Rentzsch B, Kohn B, Hoeke L,
Einspanier R (2006) Characterization of key genes of
the renin-angiotensin system in mature feline
adipocytes and during invitro adipogenesis. J Anim
Physiol Anim Nutr 100:11391148
Risch L, Huber AR (2002) Glucocorticoids and increased
serum cystatin C concentrations. Clin Chim Acta
320:133134
Rocco MV, Chen Y, Goldfarb S, Ziyadeh FN (1992)
Elevated glucose stimulates TGF-beta gene expression
and bioactivity in proximal tubule. Kidney Int 41
(1):107114
Rodríguez-Rodríguez R, González-Bulnes A, Garcia-
Contreras C et al (2020) The Iberian pig fed with
high-fat diet: a model of renal disease in obesity and
metabolic syndrome. Int J Obes 44:457465
Salim S, Hayek SS, Sever S, Ko Y-A, Tratchman H, Awad
M et al (2016) Soluble urokinase receptor and chronic
kidney disease. N Engl J Med 374(9):891
Schepers E, Barreto DV, Liabeuf S, Glorieux G, Eloot S,
Barreto FC et al (2011) Symmetric dimethylarginine as
a proinammatory agent in chronic kidney disease.
Clin J Am Soc Nephrol 6(10):23742383
Schewedhelm E, Xanthakis V, Maas R, Sullivan LM,
Atzler D, Lüneburg N et al (2011) Plasma symmetric
dimethylarginine reference limits from the
Framingham offspring cohort. Clin Chem Lab Med
49(11):19071910
Schrijvers BF, Flyvbjerg A, Tilton RG (2005) Pathophys-
iological role of vascular endothelial growth factor in
the remnant kidney. Nephron Exp Nephrol 101(1):e9
e15
Sharma AM, Engeli S (2006) Obesity and the renin
angiotensinaldosterone system. Expert Rev
Endocrinol Metab 1(2):255264
Sharma K, Ramachandrarao S, Qiu G, Usui HK, Zhu Y,
Dunn SR et al (2008) Adiponectin regulates albumin-
uria and podocyte function in mice. J Clin Invest 118
(5):16451656
Sibal L, Agarwal SC, Home PD, Boger RH (2010) The
role of asymmetric dimethylarginine (ADMA) in endo-
thelial dysfunction and cardiovascular disease. Curr
Cardiol Rev 6(2):8290
Simmons WW, Closs EI, Cunningham JM, Smith TW,
Kelly RA (1996) Cytokines and insulin induce cationic
amino acid transporter (CAT) expression in cardiac
myocytes. Regulation of L-arginine transport and no
production by CAT-1, CAT-2A, and CAT-2B. J Biol
Chem 271(2):1169411702
Singer E, Markó L, Paragas N, Barasch J, Dragun D,
Müller DN et al (2013) Neutrophil gelatinase-
associated lipocalin: pathophysiologyand clinical
applications. Acta Physiol 207(4):663672
Siroen MP, van der Sijp JR, Teerlink T, van Schaik C,
Nijveldt RJ, van Leeuwen PA (2005) The human liver
clears both asymmetric and symmetric
dimethylarginine. Hepatology 41(3):559565
Song S, Meyer M, Türk TR, Wild B, Feldkamp T, Assert
R et al (2009) Serum cystatin C in mouse models: a
reliable and precise marker for renal function and
superior to serum creatinine. Nephrol Dial Transplant
24(4):11571161
Animal Models and Renal Biomarkers of Diabetic Nephropathy
Spurlock ME, Gabler NK (2008) The development of
porcine models of obesity and the metabolic syndrome.
J Nutr 138(2):397402
Steffes MW, Buchwald H, Wigness BD (1982) Diabetic
nephropathy in the uninephrectomized dog: micro-
scopic lesions after one year. Kidney Int 21
(5):721724
Steinbach S, Weis J, Schweighauser A, Francey T, Neiger
R (2014) Plasma and urine neutrophil gelatinase
associated Lipocalin (NGAL) in dogs with acute kid-
ney injury or chronic kidney disease. J Vet Intern Med
28(2):264269
Steubl D, Block M, Herbst V (2016) Plasma uromodulin
correlates with kidney function and identies early
stages in chronic kidney disease patients. Medicine
(Baltimore) 95(10):e3011
Stevens LA, Levey AS (2009) Measured GFR as a conr-
matory test for estimated GFR. J Am Soc Nephrol 20
(11):23052313
Stevens LA, Schmid C, Greene T, Li L, Beck GJ, Joffe M
et al (2009) Factors other than GFR affecting serum
cystatin C levels. Kidney Int 75(6):652660
Straczkowski M, Kowalska I, Nikolajuk A et al (2007)
Increased serum interleukin-18 concentration is
associated with hypoadiponectinemia in obesity, inde-
pendently of insulin resistance. Int J Obes 31
(2):221225
Stridh S, Palm F, Hansell P (2012) Renal interstitial
hyaluronan- functional aspects during normal and
pathological conditions. Am J Phys Regul Integr
Comp Phys 302(11):R1235R1249
Sureshbabu A, Muhsin SA, Choi ME (2016) TGF-βsig-
naling in the kidney: probrotic and protective effects.
Am J Phys 310(7):F596F606
Theilade S, Lyngbaek S, Hansen TW, Eugen-Olsen J,
Fenger M, Rossing P et al (2015) Soluble urokinase
plasminogen activator receptor levels are elevated and
associated with complications in patients with type
1 diabetes. J Intern Med 277(3):362371
Togashi Y, Miyamoto Y (2013) Urinary cystatin C as a
biomarker for diabetic nephropathy and its immuno-
histochemical localization in kidney in Zucker diabetic
fatty (ZDF) rats. Exp Toxicol Pathol 65(5):615622
Tojo A, Onozato ML, Ha H, Kurihara H, Sakai T, Goto A
et al (2001) Reduced albumin reabsorption in the prox-
imal tubule of early-stage diabetic rats. Histochem Cell
Biol 116(3):269276
Tuttle KR, Bakris GL, Bilous RW, Coresh J, Balk E,
Kausz AT et al (2014) Diabetic kidney disease: a report
from ADA consensus conference. Diabetes Care 37
(10):28642883
Tvari-jonaviciute (2013) Effect of weight loss in obese
dogs on indicators of renal function or disease. J Vet
Intern Med 27:3138
Vallon V, Thomson SC (2012) Renal function in diabetic
disease models: the tubular system in the pathophysi-
ology of the diabetic kidney. Annu Rev Physiol
74:351375
van Hoek I, Daminet S, Notebaert S, Janssens I, Meyer E
(2008) Immunoassay of urinary retinol binding protein
as a putative renal marker in cats. J Immnunol Methods
329(12):208213
Vijay S, Hamide A, Senthilkumar GP, Mehalingam V
(2018) Urinary biomarkers for early diabetic nephrop-
athy in type 2 diabetic patients. Diabetes Metab Syndr
12(5):649652
Von Eynatten M, Lepper PM, Liu D, Lang K,
Baumann M, Nawroth PP et al (2007) Retinol-binding
protein 4 is associated with components of the meta-
bolic syndrome, but not with insulin resistance, in men
with type 2 diabetes or coronary artery disease.
Diabetologia 50(9):19391937
Wang IC, Hsu WL, Wu PH, Yin HY, Tsai HJ, Lee YJ
(2017) Neutrophil gelatinase-associated lipocalin in
cats with naturally occurring chronic kidney disease.
J Vet Intern Med 31(1):102108
Wei C, Möller CC, Altintas MM, Li J, Schwarz K,
Zacchigna S et al (2008) Modication of kidney barrier
function by the urokinase receptor. Nat Med 14
(1):5563
Williams TL, Archer J (2016) Evaluation of urinary
biomarkers for azotemic chronic disease in cats. J
Small Anim Pract 57(3):122129
Williams B, Gallacher B, Patel H, Orme C (1997)
Glucose-induced protein kinase C activation regulates
vascular permeability factor mRNA expression and
peptide production by human vascular smooth muscle
cells in vitro. Diabetes 46(9):14971503
Wiromrat P, Bjornstad P, Roncal C, Pyle L, Johnson RJ,
Cherney DZ et al (2019) Serum uromodulin is
associated with urinary albumin excretion in
adolescents with type 1 diabetes. J Diabetes Complicat
33(9):648650
Wolf G, Chen S, Zidayeh FN (2005) From the periphery of
the glomerular capillary wall toward the center of
disease: podocyte injury comes of age in diabetic
nephropathy. Diabetes 54(6):16261634
World Health Organization (WHO) (2019) Diabetes: data
and statistics. World Health Organization,
Copenhagen. [Cited 2019 May 12]. Available from:
http://www.euro.who.int/en/health-topics/
noncommunicable-diseases/diabetes/data-and-
statistics
Xu T, Sheng Z, Yao L (2017) Obesity-related
glomerulopathy- pathogenesis, pathologic, clinical
characteristics and treatment. Front Med 11(3):340348
Yacoub R, Campbell KN (2015) Inhibition of RAS in
diabetic nephropathy. Int J Nephrol Renovasc Dis
8:2940
Yang Q, Graham TE, Mody N, Preitner F, Peroni OD,
Zabolotny JM et al (2005) Serum retinol binding pro-
tein 4 contributes to insulin resistance in obesity and
type 2 diabetes. Nature 436(7049):356362
Zeni L, Norden AGW, Cancarini G, Unwin RJ (2017) A
more tubulocentric view of diabetic kidney disease. J
Nephrol 30(6):701717
L. Pérez-López et al.
Zhang X, Lerman LO (2016) Investigating the metabolic
syndrome: contributions of swine models. Toxicol
Pathol 44(3):358366
Zini E, Benali S, Coppola L, Guscetti F, Ackermann M,
Lutz TA et al (2014) Renal morphology in cats with
diabetes mellitus. Vet Pathol 51(6):11431150
Ziyadeh FN (2004) Mediators of diabetic renal disease: the
case for tgf-beta as the major mediator. J Am Soc
Nephrol 15(Suppl1):S55S57
Ziyadeh FN, Hoffman BB, Han DC, Iglesias-De La Cruz
MC, Hong SW, Isono M et al (2000) Long-term pre-
vention of renal insufciency, excess matrix gene
expression, and glomerular mesangial matrix expan-
sion by treatment with monoclonal antitransforming
growth factor-beta antibody in db/db diabetic mice.
Proc Nat Acad Sci U S A 97(14):80158020
Zobel EH, von Scholten BJ, Reinhard H (2017) Symmetric
and asymmetric dimethylarginine as risk markers of
cardiovascular disease, all-cause mortality and deterio-
ration in kidney function in persons with type 2 diabe-
tes and microalbuminuria. Cardiovasc Diabetol 16
(4):888896
Zsuga J, Török J, Magyar MT, Valikovics A, Gesztelyi R,
Lenkei A et al (2007) Dimethylarginines at the cross-
road of insulin resistance and aterosclerosis. Metabo-
lism 56(3):394300
Animal Models and Renal Biomarkers of Diabetic Nephropathy
... Blood urea nitrogen, GFR estimation formula, serum creatinine, and albuminuria are currently utilized to evaluate the presence and progression of diabetic nephropathy (DN). However, they lack precision and sensitivity towards minor changes in renal function (14,15). The most dependable indicator for renal function in patients with autosomal dominant polycystic kidney disease (ADPKD) is the height corrected total kidney volume (htTKV). ...
Article
Full-text available
The Monocyte chemoattractant protein-1 (MCP-1), also referred to as chemokine ligand 2 (CCL2), belongs to the extensive chemokine family and serves as a crucial mediator of innate immunity and tissue inflammation. It has a notable impact on inflammatory conditions affecting the kidneys. Upon binding to its receptor, MCP-1 can induce lymphocytes and NK cells’ homing, migration, activation, differentiation, and development while promoting monocytes’ and macrophages’ infiltration, thereby facilitating kidney disease-related inflammation. As a biomarker for kidney disease, MCP-1 has made notable advancements in primary kidney diseases such as crescentic glomerulonephritis, chronic glomerulonephritis, primary glomerulopathy, idiopathic proteinuria glomerulopathy, acute kidney injury; secondary kidney diseases like diabetic nephropathy and lupus nephritis; hereditary kidney diseases including autosomal dominant polycystic kidney disease and sickle cell kidney disease. MCP-1 not only predicts the occurrence, progression, prognosis of the disease but is also closely associated with the severity and stage of nephropathy. When renal tissue is stimulated or experiences significant damage, the expression of MCP-1 increases, demonstrating a direct correlation with the severity of renal injury.
... However, their primitive form of renal cells is functionally not similar to humans and these features need to be replicated in more advanced models, although, these models can be utilized for genetic or drug screening. Companion animals such as cats and dogs are also studied as they develop diabetes and share the same environment [151]. Obese cats have exhibited many alterations associated with metabolic syndrome in people, despite these changes hypertension, atherosclerosis or association with renal disease is unclear [152,153]. ...
Article
Full-text available
Introduction Diabetic nephropathy (DN), an outcome of prolonged diabetes, has affected millions of people worldwide and every year the incidence and prevalence increase substantially. The symptoms may start with mild manifestations of the disease such as increased albuminuria, serum creatinine levels, thickening of glomerular basement membrane, expansion of mesangial matrix to severe pathological symptoms such as glomerular lesions and tubulointerstitial fibrosis which may further proceed to cardiovascular dysfunction or end-stage renal disease. Perspective Numerous therapeutic interventions are being explored for the management of DN, however, these interventions do not completely halt the progression of this disease and hence animal models are being explored to identify critical genetic and molecular parameters which could help in tackling the disease. Rodent models which mostly include mice and rats are commonly used experimental animals which provide a wide range of advantages in understanding the onset and progression of disease in humans and also their response to a wide range of interventions helps in the development of effective therapeutics. Rodent models of type 1 and type 2 diabetes induced DN have been developed utilizing different platforms and interventions during the last few decades some of which mimic various stages of diabetes ranging from early to later stages. However, a rodent model which replicates all the features of human DN is still lacking. This review tries to evaluate the rodent models that are currently available and understand their features and limitations which may help in further development of more robust models of human DN. Conclusion Using these rodent models can help to understand different aspects of human DN although further research is required to develop more robust models utilizing diverse genetic platforms which may, in turn, assist in developing effective interventions to target the disease at different levels.
... Studies have shown that VASH-1 has a protective effect on renal function in patients with diabetic nephropathy. 10 Therefore, this study explored the differences and clinical significance of serum 25-hydroxyvitamin D3 and VASH-1 levels in patients with diabetic nephropathy and different renal injuries. ...
Article
Full-text available
Objective We investigate the relationship between the changes of serum 25-hydroxyvitamin D3 (25(OH)D3) and vasohibin-1 (VASH-1) and renal function injury in patients with type 2 diabetic nephropathy. Methods In this study, 143 patients with diabetic nephropathy (DN) were selected as DN group, and 80 patients with type 2 diabetes mellitus were selected as T2DM group. The serum 25 (OH) D3, VASH-1, blood glucose index, inflammation index and renal function index were compared between the two groups. According to the urinary microalbumin/creatinine ratio (UACR), the DN group was divided into microalbuminuria group (UACR range≥30.0mg/g and <300.0mg/g) and macroalbuminuria group (UACR≥300.0mg/g) for stratified comparison. The correlation between 25-hydroxyvitamin D3, VASH-1 and inflammation index and renal function index was analyzed by simple linear correlation analysis. Results The level of 25 (OH) D3 in DN group was significantly lower than that in T2DM group (P<0.05). The levels of VASH-1, CysC, BUN, Scr, 24h urine protein, serum CRP, TGF-β1, TNF-α and IL-6 in DN group were higher than those in T2DM group (P<0.05). The level of 25 (OH) D3 in DN patients with massive proteinuria was significantly lower than that in DN patients with microalbuminuria. The level of VASH-1 in DN patients with massive proteinuria was higher than that in DN patients with microalbuminuria (P<0.05). There was a negative correlation between 25 (OH) D3 and CysC, BUN, Scr, 24h urine protein, CRP, TGF-β1, TNF-α, IL-6 in patients with DN (P<0.05). VASH-1 was positively correlated with Scr, 24h urinary protein, CRP, TGF-β1, TNF-α and IL-6 in patients with DN (P<0.05). Conclusion The level of serum 25 (OH) D3 in DN patients was considerably decreased, and the level of VASH-1 was increased, which was related to the degree of renal function injury and inflammatory response.
... The primary clinical symptom of DN is progressive albuminuria [5,6], but the underlying mechanism still needs to be elucidated. Despite recent advances in DN research, there has been no breakthrough in developing reliable early diagnostic markers or effective treatments for this devastating disease [7][8][9][10]. Therefore, there is an unmet medical need to understand the pathogenesis of DN better for facilitating the discovery and development of new targeted therapies. ...
Article
Full-text available
Progressive albuminuria is the primary clinical symptom of diabetic nephropathy (DN), leading to a gradual decline in kidney function. DLX6-AS1 was the first reported long non-coding RNA (lncRNA) to participate in organogenesis and play crucial roles in the brain or neural cell development. Herein, we investigated the DLX6-AS1 (Dlx6-os1 in mice) role in DN pathogenesis. We found that DLX6-AS1 expression in DN patients correlated with the extent of albuminuria. Dlx6-os1 overexpression induced cellular damage and inflammatory responses in cultured podocytes through miR-346-mediated regulation of the GSK-3β pathway. In various established diabetic and newly developed knockout mouse models, Dlx6-os1 knockdown/knockout significantly reduced podocyte injury and albuminuria. The Dlx6-os1 effects were remarkably modulated by miR-346 mimics or mutants and significantly diminished in podocyte-specific GSK-3β-knockout mice. Thus, DLX6-AS1 (Dlx6-os1) promotes DN development by accelerating podocyte injury and inflammation through the upregulation of the GSK-3β pathway, providing a novel molecular target for DN therapy.
... e early clinical diagnosis of DN is based on canonical biochemical markers, such as glomerular filtration rate, urinary microalbumin, urinary microalbumin to urinary creatinine ratio, serum creatinine, urinary cystatin C, and serum β2 microglobulin. In addition, some biomarkers related to the pathogenesis of DN, such as kidney injury molecule 1 (Kim-1), neutrophil gelatinase-associated lipocalin (NGAL), tissue inhibitor of metalloproteinases-2, insulin-like growth factorbinding protein 7 (IGFBP-7), vascular endothelial growth factor (VEGF), transforming growth factor-β (TGF-β), monocyte chemoattractant protein-1(MCP-1), and inflammatory cytokines, such as tumor necrosis factor (TNF)α, MCP-1, and ILs (IL-1α, IL-1β, IL-18, IL-10), have also attracted significant attention [24,25]. However, the pathogenesis of DN remains unclear, and treatment strategies are limited. ...
Article
Full-text available
Pyroptosis is an inflammatory form of programmed cell death that is dependent on inflammatory caspases, leading to the cleavage of gasdermin D (GSDMD) and increased secretion of interleukin (IL)-1β and IL-18. Recent studies have reported that hyperglycemia-induced cellular stress stimulates pyroptosis, and different signaling pathways have been shown to play crucial roles in regulating pyroptosis. This review summarized and discussed the molecular mechanisms, regulation, and cellular effects of pyroptosis in diabetic microvascular complications, such as diabetic nephropathy, diabetic retinopathy, and diabetic cardiomyopathy. In addition, this review aimed to provide new insights into identifying better treatments for diabetic microvascular complications.
... Animal experiments published to assess biomarkers and their association with kidney diseases or DN[15]. ...
Article
Full-text available
The inflammatory component of diabetic kidney disease has become of great interest in recent years, with genetic and epigenetic variants playing a fundamental role in the initiation and progression of the disease. Cells of the innate immune system play a major role in the pathogenesis of diabetic kidney disease, with a lesser contribution from the adaptive immune cells. Other components such as the complement system also play a role, as well as specific cytokines and chemokines. The inflammatory component of diabetic kidney disease is of great interest and is an active research field, with the hope to find potential innovative therapeutic targets.
Article
Full-text available
Background There are no reliable molecular targets for early diagnosis and effective treatment in the clinical management of diabetic kidney disease (DKD). To identify novel gene factors underlying the progression of DKD. Methods The public transcriptomic datasets of the alloxan-induced DKD model and the streptozotocin-induced DKD model were retrieved to perform an integrative bioinformatic analysis of differentially expressed genes (DEGs) shared by two experimental animal models. The dominant biological processes and pathways associated with DEGs were identified through enrichment analysis. The expression changes of the key DEGs were validated in the classic db/db DKD mouse model. Results The downregulated and upregulated genes in DKD models were uncovered from GSE139317 and GSE131221 microarray datasets. Enrichment analysis revealed that metabolic process, extracellular exosomes, and hydrolase activity are shared biological processes and molecular activity is altered in the DEGs. Importantly, Hmgcs2, angptl4, and Slco1a1 displayed a consistent expression pattern across the two DKD models. In the classic db/db DKD mice, Hmgcs2 and angptl4 were also found to be upregulated while Slco1a1 was downregulated in comparison to the control animals. Conclusions In summary, we identified the common biological processes and molecular activity being altered in two DKD experimental models, as well as the novel gene factors (Hmgcs2, Angptl4, and Slco1a1) which may be implicated in DKD. Future works are warranted to decipher the biological role of these genes in the pathogenesis of DKD.
Article
Diabetic nephropathy is one of the most common complications in diabetes. It has been shown to be the leading cause of end-stage renal disease. However, due to their complex pathological mechanisms, effective therapeutic drugs other than angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs), which have been used for 20 years, have not been developed so far. Recent studies have shown that diabetic nephropathy is characterized by multiple signalling pathways and multiple targets, including inflammation, apoptosis, pyroptosis, autophagy, oxidative stress, endoplasmic reticulum stress and their interactions. It definitely exacerbates the difficulty of therapy, but at the same time it also brings out the chance for natural products treatment. In the most recent two decades, a large number of natural products have displayed their potential in preclinical studies and a few compounds are under invetigation in clinical trials. Hence, many compounds targeting these singals have been emerged as a comprehensive blueprint for treating strategy of diabetic nephropathy. This review focuses on the cellular and molecular mechanisms of natural prouducts that alleviate this condition, including preclinical studies and clinical trials, which will provide new insights into the treatment of diabetic nephropathy and suggest novel ideas for new drug development.
Article
One of the most important steps in drug discovery is the translation of preclinical data to humans. However, the term 'translation' has numerous connotations and, often, different stakeholders literally speak different languages. Learning from many years of experience and new concepts in language translation could increase the success rate in translating biomedical research. Beyond being bilingual, this includes applying the concept of functional equivalence, the main characteristic of a good translation. Given that function is defined by the source language text, starting with the patient has advantages over the classical bench-to-bedside approach. Good translators need transfer competence, including knowledge of the limitations of translation. As with languages, computer-assisted translation(-al research) could support increasing functional equivalence and, thus, translation success.
Article
Diabetic nephropathy (DN) is a common complication of diabetes and it is urgent to develop effective therapies for DN. In this study, high‐sucrose and high‐fat diet combined with streptozotocin was used to induce DN in rats to observe the effects of natural flavonoid ombuin on renal function, inflammation, and interstitial fibrosis. Immunohistochemistry and western blotting analysis were used to detect protein expression levels. Results showed that ombuin significantly improved renal function and pathological injury, inhibited accumulation of advanced glycation end‐products, suppressed the release of inflammatory cytokines, and improved renal interstitial fibrosis in DN rats. Ombuin also significantly downregulated the expressions of transforming growth factor beta1 (TGF‐β1), connective tissue growth factor (CTGF), fibronectin (FN), p65, phosphorylated (p)‐p65, Cleaved‐Notch 1, and hairy and enhancer of split 1 (Hes 1), and upregulated the expression of peroxisome proliferator‐activated receptor γ (PPAR γ). When PPAR γ activity was inhibited by T0070907, the effects of ombuin on improving DN were significantly reversed, and the expressions of TGF‐β1, FN, CTGF, p‐p65, and p65 increased, while the expressions of Cleaved‐Notch 1 and Hes 1 were not significantly affected. These results suggest that ombuin may activate PPAR γ to exert anti‐inflammatory and antifibrotic effects by inhibiting Notch 1 activity in DN. It is also possible that ombuin acts on these two independent signal pathways synchronously.
Article
Full-text available
The pathogenesis of renal disease in the context of overweight/obesity, metabolic syndrome, and insulin resistance is not completely understood. This may be due to the lack of a definitive animal model of disease, which limits our understanding of obesity-induced renal damage. We evaluated the changes in renal histology and lipid deposits induced by obesity in a model of insulin resistance: the Iberian swine fed with fat-enriched food. Twenty-eight female sows were randomized to standard (SD) or high-fat diet (HFD: 6.8% of saturated fat) for 100 days. Weight, adiposity, analytics, oral glucose tolerance tests, and measured renal function were determined. Renal histology and lipid deposits in renal tissue were analyzed. Animals on HFD developed obesity, hypertension, high levels of LDL cholesterol, triglycerides, insulin resistance, and glomerular hyperfiltration. No animal developed overt diabetes. Animals on HFD showed “diabetoid changes”, including mesangial expansion [21.40% ± 4 vs.13.20% ± 4.0, p < 0.0001], nodular glomerulosclerosis [7.40% ± 7, 0.75 vs. 2.40% ± 4.7, p = 0.02], and glomerulomegaly (18% vs. 10%, p = 0.010) than those on SD. Tubular atrophy, interstitial fibrosis, inflammation, arteriolar hyalinosis, or fibrointimal thickening were mild and similar between groups. Triglyceride content in renal tissue was higher in animals on HFD than in SD (15.4% ± 0.5 vs. 12.7% ± 0.7; p < 0.01). Iberian pigs fed with fat-enriched food showed diabetoid changes and glomerulomegaly as observed in obese humans making this model suitable to study obesity-induced renal disease.
Article
Full-text available
Background Diabetes mellitus is the main cause of chronic kidney disease (CKD) in humans. The relationship between the 2 diseases in cats is unclear. Objective To assess the association between diabetes and CKD in a population of adult cats. Animals Five hundred sixty‐one cats that attended 2 veterinary centers in Gran Canaria, Spain, between 2014 and 2016. Methods Medical records were retrospectively reviewed. Cats aged 3 years or older, with sufficient data to define whether or not they had diabetes and CKD, were selected. Cats in critical condition, with dehydration or potential causes of prerenal azotemia and those treated with nephrotoxic drugs were excluded. Diagnosis of CKD was established when creatinine concentrations were >2 mg/dL, or serum creatinine 1.6‐2 mg/dL and urine specific gravity <1.035, or serum creatinine 1.6‐2 mg/dL and urine protein/creatinine ratio >0.4. Factors associated with CKD were identified through multivariate logistic regression analyses. Results Sixty‐seven (11.9%) cats had CKD and 16 (2.9%) cats had diabetes. Sixty cats without diabetes (11%) and 7 with diabetes (44%) had CKD. Among the latter, both conditions were diagnosed simultaneously in 6 cases, whereas diabetes preceded CKD in the other. Multivariate analysis showed that diabetes was significantly associated with CKD (odds ratio = 4.47; 95% confidence interval, 1.51‐13.28; P = .007). Other variables associated with CKD were age and mixed breed. Conclusions and Clinical Importance After adjusting for age, this study showed an association between diabetes and CKD in adult cats.
Article
Full-text available
Aims/hypothesis We aimed to identify a sparse panel of biomarkers for improving the prediction of renal disease progression in type 1 diabetes. Methods We considered 859 individuals recruited from the Scottish Diabetes Research Network Type 1 Bioresource (SDRNT1BIO) and 315 individuals from the Finnish Diabetic Nephropathy (FinnDiane) study. All had an entry eGFR between 30 and 75 ml min⁻¹[1.73 m]⁻², with those from FinnDiane being oversampled for albuminuria. A total of 297 circulating biomarkers (30 proteins, 121 metabolites, 146 tryptic peptides) were measured in non-fasting serum samples using the Luminex platform and LC electrospray tandem MS (LC-MS/MS). We investigated associations with final eGFR adjusted for baseline eGFR and with rapid progression (a loss of more than 3 ml min⁻¹[1.73 m]⁻² year⁻¹) using linear and logistic regression models. Panels of biomarkers were identified using a penalised Bayesian approach, and their performance was evaluated through 10-fold cross-validation and compared with using clinical record data alone. Results For final eGFR, 16 proteins and 30 metabolites or tryptic peptides showed significant association in SDRNT1BIO, and nine proteins and five metabolites or tryptic peptides in FinnDiane, beyond age, sex, diabetes duration, study day eGFR and length of follow-up (all at p < 10⁻⁴). The strongest associations were with CD27 antigen (CD27), kidney injury molecule 1 (KIM-1) and α1-microglobulin. Including the Luminex biomarkers on top of baseline covariates increased the r² for prediction of final eGFR from 0.47 to 0.58 in SDRNT1BIO and from 0.33 to 0.48 in FinnDiane. At least 75% of the increment in r² was attributable to CD27 and KIM-1. However, using the weighted average of historical eGFR gave similar performance to biomarkers. The LC-MS/MS platform performed less well. Conclusions/interpretation Among a large set of associated biomarkers, a sparse panel of just CD27 and KIM-1 contains most of the predictive information for eGFR progression. The increment in prediction beyond clinical data was modest but potentially useful for oversampling individuals with rapid disease progression into clinical trials, especially where there is little information on prior eGFR trajectories.
Article
Full-text available
Since inbred C57BL/6 mice are known to show inter-individual phenotypic variability for some traits, we tested the hypothesis that inbred C57BL/6 mice display a different tendency to consume a high fat (HF) diet. For this purpose, we used a compilation of HF intake data from an experimental protocol in which satiated mice were exposed to a HF pellet every morning for 2-h over 4 consecutive days. We found that mice displayed a large degree of variability in HF intake. Since day 1 HF intake significantly correlated with HF intake in successive days, we applied a hierarchical clustering algorithm on HF intake measurements in days 2, 3, and 4 in order to classify mice into “low” or “high” HF intake groups. “Low” HF intake group showed a day 1 HF intake similar to that seen in mice exposed to regular chow, while “high” HF intake group showed a higher day 1 HF intake as compared to “low” HF intake group. Both groups of mice increased HF consumption over the successive days, but “high” HF intake group always displayed a higher HF consumption than the “low” HF intake group. As compared to “low” HF intake group, “high” HF intake group showed a higher number of dopamine neurons positive for c-Fos in the VTA after the last event of HF intake. Thus, inbred C57BL/6 mice show inter-individual variability for HF intake and such feature may be linked to a different response to the rewarding properties of the HF diet.
Article
Full-text available
Objective: Soluble urokinase plasminogen activator receptor (suPAR) is an important inflammatory biomarker implicated in endothelial and podocyte dysfunction. However, suPAR's predictive qualities for complications in type 1 diabetes have yet to be determined. We investigated the prognostic value of suPAR for the development of cardiovascular events, decline in renal function, and mortality in patients with type 1 diabetes. Research design and methods: We included 667 patients with type 1 diabetes with various degrees of albuminuria in a prospective study. End points were cardiovascular events (cardiovascular death, nonfatal acute myocardial infarction, nonfatal stroke, or coronary or peripheral arterial interventions), estimated glomerular filtration rate (eGFR) decline ≥30%, progression from lower to higher albuminuric state, development of end-stage renal disease (ESRD), and mortality. Follow-up was 5.2-6.2 years. Results were adjusted for known risk factors. Hazard ratios (HRs) are presented per doubling of suPAR with 95% CI. Relative integrated discrimination improvement (rIDI) was calculated. Results: Quantification of suPAR was available in all participants; median (interquartile range) was 3.4 ng/mL (2.7-4.5). The adjusted HR (95% CI) for cardiovascular events (n = 94), progression in albuminuria (n = 36), eGFR decline (n = 93), ESRD (n = 23), and mortality (n = 58) were 3.13 (1.96-5.45, P < 0.001), 1.27 (0.51-3.19, P = 0.61), 2.93 (1.68-5.11, P < 0.001), 2.82 (0.73-11.9, P = 0.13), and 4.13 (1.96-8.69, P < 0.001), respectively. rIDI was significant for cardiovascular events (22.6%, P < 0.001), eGFR decline (14.4%, P < 0.001), and mortality (23.9%, P < 0.001). Conclusions: In patients with type 1 diabetes and a broad range of albuminuria, a higher level of suPAR is a significant and independent risk factor for cardiovascular events, decline in eGFR ≥30%, and mortality. In addition, suPAR contributes significantly to discrimination for the end points.
Article
Full-text available
Background Early detection of decreased glomerular filtration rate (GFR) in dogs is challenging. Current methods are insensitive and new biomarkers are required. Objective To compare overall diagnostic performance of serum symmetric dimethylarginine (SDMA) and serum cystatin C to serum creatinine, for detection of decreased GFR in clinically stable dogs, with or without chronic kidney disease (CKD). Animals Ninety‐seven client‐owned dogs: 67 dogs with a diagnosis or suspicion of CKD and 30 healthy dogs were prospectively included. Methods Prospective diagnostic accuracy study. All dogs underwent physical examination, systemic arterial blood pressure measurement, urinalysis, hematology and blood biochemistry analysis, cardiac and urinary ultrasound examinations, and scintigraphy for estimation of glomerular filtration rate (mGFR). Frozen serum was used for batch analysis of SDMA and cystatin C. Results The area under the curve of creatinine, SDMA, and cystatin C for detection of an mGFR <30.8 mL/min/L was 0.98 (95% confidence interval [CI], 0.93‐1.0), 0.96 (95% CI, 0.91‐0.99), and 0.87 (95% CI, 0.79‐0.93), respectively. The sensitivity of both creatinine and SDMA at their prespecified cutoffs (115 μmol/L [1.3 mg/dL] and 14 μg/dL) for detection of an abnormal mGFR was 90%. The specificity was 90% for creatinine and 87% for SDMA. When adjusting the cutoff for cystatin C to correspond to a diagnostic sensitivity of 90% (0.49 mg/L), specificity was lower (72%) than that of creatinine and SDMA. Conclusions and Clinical Importance Overall diagnostic performance of creatinine and SDMA for detection of decreased mGFR was similar. Overall diagnostic performance of cystatin C was inferior to both creatinine and SDMA.
Article
Full-text available
Diseases of the kidney are difficult to diagnose and treat. This review summarises the definition, cause, epidemiology and treatment of some of these diseases including chronic kidney disease, diabetic nephropathy, acute kidney injury, kidney cancer, kidney transplantation and polycystic kidney diseases. Numerous studies have adopted a metabolomics approach to uncover new small molecule biomarkers of kidney diseases to improve specificity and sensitivity of diagnosis and to uncover biochemical mechanisms that may elucidate the cause and progression of these diseases. This work includes a description of mass spectrometry-based metabolomics approaches, including some of the currently available tools, and emphasises findings from metabolomics studies of kidney diseases. We have included a varied selection of studies (disease, model, sample number, analytical platform) and focused on metabolites which were commonly reported as discriminating features between kidney disease and a control. These metabolites are likely to be robust indicators of kidney disease processes, and therefore potential biomarkers, warranting further investigation.
Article
. Impaired angiogenesis and decreased vascular endothelial growth factor (VEGF) expression were recently documented in the remnant kidney (RK) model of progressive renal failure. VEGF (50 μg/kg, twice daily) was administered to RK rats between weeks 4 and 8 after surgery, and rats were euthanized at week 8 for histologic study. During the administration of VEGF ( n = 7) or vehicle ( n = 6), systemic BP was comparable in the two groups. VEGF treatment resulted in improved renal function and lower mortality rates, compared with the vehicle-treated group. Renal histologic analyses confirmed a 3.5-fold increase in glomerular endothelial cell proliferation (0.14 ± 0.03 versus 0.04 ± 0.02 proliferating endothelial cells/glomerulus, VEGF versus vehicle, P < 0.05), a twofold increase in peritubular capillary endothelial cell proliferation (1.60 ± 0.30 versus 0.78 ± 0.17 cells/mm ² , VEGF versus vehicle, P < 0.01), a threefold decrease in peritubular capillary rarefaction ( P < 0.01), and a twofold increase in endothelial nitrix oxide synthase expression ( P < 0.05) in the VEGF-treated group; an eightfold increase in urinary nitrate/nitrite levels ( P < 0.05) was also noted. Although the difference in glomerulosclerosis scores did not reach statistical significance (0.67 ± 0.42 versus 1.22 ± 0.63, VEGF versus vehicle; range, 0 to 4; P = NS), VEGF-treated rats exhibited less interstitial collagen type III deposition (9.32 ± 3.26 versus 17.45 ± 7.50%, VEGF versus vehicle, P < 0.01) and reduced tubular epithelial cell injury, as manifested by osteopontin expression (5.57 ± 1.60 versus 9.58 ± 3.45%, VEGF versus vehicle, P < 0.01). In conclusion, VEGF treatment reduces fibrosis and stabilizes renal function in the RK model. The use of angiogenic factors may represent a new approach to the treatment of kidney disease.
Article
Clinical risk factors explain only a fraction of the variability of estimated glomerular filtration rate (eGFR) decline in people with type 2 diabetes. Cross-omics technologies by virtue of a wide spectrum screening of plasma samples have the potential to identify biomarkers for the refinement of prognosis in addition to clinical variables. Here we utilized proteomics, metabolomics and lipidomics panel assay measurements in baseline plasma samples from the multinational PROVALID study (PROspective cohort study in patients with type 2 diabetes mellitus for VALIDation of biomarkers) of patients with incident or early chronic kidney disease (median follow-up 35 months, median baseline eGFR 84 mL/min/1.73 m2, urine albumin-to-creatinine ratio 8.1 mg/g). In an accelerated case-control study, 258 individuals with a stable eGFR course (median eGFR change 0.1 mL/min/year) were compared to 223 individuals with a rapid eGFR decline (median eGFR decline -6.75 mL/min/year) using Bayesian multivariable logistic regression models to assess the discrimination of eGFR trajectories. The analysis included 402 candidate predictors and showed two protein markers (KIM-1, NTproBNP) to be relevant predictors of the eGFR trajectory with baseline eGFR being an important clinical covariate. The inclusion of metabolomic and lipidomic platforms did not improve discrimination substantially. Predictions using all available variables were statistically indistinguishable from predictions using only KIM-1 and baseline eGFR (area under the receiver operating characteristic curve 0.63). Thus, the discrimination of eGFR trajectories in patients with incident or early diabetic kidney disease and maintained baseline eGFR was modest and the protein marker KIM-1 was the most important predictor.
Article
Early diabetic kidney disease (DKD) occurs in adolescents with type 1 diabetes (T1D). Lower serum uromodulin (SUMOD) predicts DKD progression in adults with T1D. In this study, we demonstrate that lower SUMOD is associated with urinary albumin excretion in adolescents with T1D, suggesting a potential relationship between SUMOD and early kidney dysfunction in T1D youth.