You are on page 1of 19

REVIEWS

Obesity-related glomerulopathy:
clinical and pathologic characteristics
and pathogenesis
Vivette D. D’Agati1, Avry Chagnac2, Aiko P.J. de Vries3, Moshe Levi4, Esteban Porrini5,
Michal Herman-Edelstein6 and Manuel Praga7
Abstract | The prevalence of obesity-related glomerulopathy is increasing in parallel with
the worldwide obesity epidemic. Glomerular hypertrophy and adaptive focal segmental
glomerulosclerosis define the condition pathologically. The glomerulus enlarges in response to
obesity-induced increases in glomerular filtration rate, renal plasma flow, filtration fraction and
tubular sodium reabsorption. Normal insulin/phosphatidylinositol 3‑kinase/Akt and mTOR
signalling are critical for podocyte hypertrophy and adaptation. Adipokines and ectopic lipid
accumulation in the kidney promote insulin resistance of podocytes and maladaptive responses
to cope with the mechanical forces of renal hyperfiltration. Although most patients have stable or
slowly progressive proteinuria, up to one-third develop progressive renal failure and end-stage
renal disease. Renin–angiotensin–aldosterone blockade is effective in the short-term but weight
loss by hypocaloric diet or bariatric surgery has induced more consistent and dramatic
antiproteinuric effects and reversal of hyperfiltration. Altered fatty acid and cholesterol
metabolism are increasingly recognized as key mediators of renal lipid accumulation,
inflammation, oxidative stress and fibrosis. Newer therapies directed to lipid metabolism,
including SREBP antagonists, PPARα agonists, FXR and TGR5 agonists, and LXR agonists, hold
therapeutic promise.

Obesity and diabetes mellitus occurring in the context Pathology of ORG


of obesity — known as diabesity — are now leading Biopsy incidence
causes of chronic kidney disease (CKD)1–8. Between 1978 The obesity epidemic has led to an increase in the num-
and 2013, the proportion of overweight and obese adults ber of renal biopsies performed in obese patients. Early
(BMI ≥25 kg/m2) worldwide increased from 28.8% to case reports of ORG were limited to autopsy studies13,
36.9% among men, and from 29.8% to 38.0% among which identified an association between extreme obesity
women9. In the USA, the prevalence of obesity (BMI and the development of glomerular hypertrophy (glo-
≥30 kg/m2) among adults aged 20–74 years has more merulomegaly). In 1974, an association between mas-
than doubled over the past three decades, from 15% to sive obesity and nephrotic-range proteinuria was first
35% in 2011–2012 (REF. 10). This increase occurred in described14. The full spectrum of ORG emerged from
men and women of all age groups and across diverse detailed clinical–pathologic studies15–17, and from stud-
ethnicities. Estimates suggest that by 2030 more than ies that contrasted ORG with primary focal segmental
50% of the US population will be obese and either at glomerulosclerosis (FSGS)16. The frequency of ORG
risk of, or experiencing, obesity-related complications11, has increased in US, European and Asian cohorts over
resulting in a substantial economic burden12. The obesity the past 30 years15–17. A retrospective study that evalu-
epidemic has led to an increased incidence of obesity- ated native kidney biopsy samples received at Columbia
related glomerulopathy (ORG), a distinct entity featuring University, New York, USA, reported a 10‑fold increase
Correspondence to proteinuria, glomerulomegaly, progressive glomerulo­ in the incidence of ORG from 0.2% in 1986–1990 to
V.D.D. vdd1@columbia.edu sclerosis and renal functional decline. Here we review 2.0% in 1996–2000 (REF. 16), with a further rise to 2.7% in
doi:10.1038/nrneph.2016.75 the pathologic and clinical features of ORG as well as the 2001–2015 (V. D’Agati, unpublished data). In this study
Published online 6 June 2016 pathogenesis and potential therapeutic targets. all ORG biopsies were performed for overt renal disease:

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 1


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Key points segmental sclerosis typically affect hypertrophied glo-


meruli (FIG. 1). They are often perihilar (contiguous with
• The incidence of obesity-related glomerulopathy (ORG) is increasing in parallel the glomerular vascular pole) but might involve any
with the worldwide obesity epidemic portion of the glomerular globe. In the Columbia study,
• Pathologic features of ORG include glomerulomegaly and focal segmental exclusively perihilar lesions were found in 19% and
glomerulosclerosis (FSGS), particularly the perihilar variant; the degree of foot admixture of perihilar and peripheral lesions in 81% of
process effacement in ORG is usually less than in primary FSGS ORG biopsy samples16. The percentage of glomeruli that
• Subnephrotic proteinuria is the most common clinical presentation of ORG; some are affected by segmental sclerosis tends to be lower in
patients have nephrotic-range proteinuria and progressive loss of renal function but ORG (range 3–50%; mean 12%) than in primary FSGS
full nephrotic syndrome is highly unusual (mean 39%), supporting the view that ORG is a milder,
• Major renal physiologic responses to obesity include increases in glomerular filtration more indolent form of FSGS16. In a Chinese cohort of
rate, renal plasma flow, filtration fraction and tubular reabsorption of sodium patients with ORG, the mean percentage of glomeruli
• Adipokines and ectopic lipid accumulation in the kidney promote maladaptive with FSGS was as low as 6%15.
responses of renal cells to the mechanical forces of hyperfiltration, leading to Among the five histologic subtypes of FSGS (not
podocyte depletion, proteinuria, FSGS and interstitial fibrosis other­wise specified, perihilar, cellular, tip and collaps-
• Therapeutic interventions include renin–angiotensin–aldosterone inhibition and ing), ORG exhibits a predominance of the perihilar
weight loss; novel strategies involve administration of small molecules that variant 20. This predilection for perihilar sclerosis might
specifically modulate deleterious pathways of fatty acid and cholesterol metabolism
reflect the greater ultrafiltration pressure (that is, the
difference between glomerular capillary hydrostatic and
oncotic pressure) at the afferent end than at the efferent
56% of patients had proteinuria and 44% had protein­uria end of the glomerular capillary bed. Such differences are
and renal insufficiency. The criteria for a diagnosis of likely exaggerated under conditions of increased filtra-
ORG were BMI ≥30 kg/m2 and the presence of glomerulo­ tion demand and reflex dilatation of the afferent arteri-
megaly with or without FSGS. The mean BMI of patients ole21. Correspondingly, ORG biopsy samples may show
with ORG was 41.7 kg/m2 (range 30.9–62.7 kg/m2); increased luminal diameter of the afferent arteriole and
46% had class 1 or class 2 obesity and 54% had class 3 the glomerular capillaries22. As the glomeruli enlarge, the
obesity (BOX 1). This study, therefore, made the important numerical density of podocytes decreases. In one study,
observation that ORG is not restricted to patients with podocyte density was 55% lower (134 per 106 μm2 versus
morbid (class 3) obesity 16. 245 per 106 μm2) and mean glomerular volume was 158%
higher (4.64 × 106 μm2 versus 2.94 × 106 μm2) in patients
Histology with ORG than in non-obese kidney donors22. Reduced
Glomerulomegaly and FSGS. Identification of glomer- podocyte density in this context might reflect both adap-
ulomegaly requires measurement of the diameters of tive hypertrophic responses to glomerulomegaly and
all glomeruli sampled or of those sectioned through the ongoing podocyte depletion that has not yet reached a
hilus — the epicentre of the glomerular globe. Other threshold for the development of segmental sclerosis23,24.
stereo­logical methods estimate glomerular volume using In experimental models of ORG, such as in Fischer
serial sections of individual glomeruli18. In the Columbia rats fed an ad libitum diet, glomerular tuft volume
series, mean glomerular diameter in biopsy samples from increases exponentially with body weight 25. Podocyte
patients with ORG was 226 μm versus 169 μm in age and cellular volume also increases in relation to body weight
sex-matched normal control samples16. Across all age gain, consistent with adaptive podocyte hypertrophy,
groups (from the first to the ninth decade of life), glo- but at a lower rate than the increase in glomerular tuft
merular diameter in patients with ORG was on average volume, indicating mismatch between these two varia-
1.34‑fold greater than that of non-obese normal controls. bles25. As podocytes cannot proliferate and their ability
FSGS is defined as a segmental consolidation of the to hypertrophy is limited, the mechanical strain on these
glomerular tuft by extracellular matrix and/or hyaline, cells resulting from stretch tension and shear stress as
causing capillary obliteration19. In ORG, lesions of glomerular volume expands, reaches a breakpoint.

Author addresses
1
Department of Pathology, Columbia University Medical Center, 630 W. 168th Street, Room VC14‑224, New York, New York 10032, USA.
2
Department of Nephrology and Hypertension, Rabin Medical Center, 39 Jabotinsky Street, 4941492, Petah Tikva, Israel and Sackler
School of Medicine, Tel Aviv University, Tel Aviv, Israel.
3
Department of Medicine, Division of Nephrology, Leiden University Medical Center and Leiden University, Room C7‑036, PO Box
9600, 2300 RC, Leiden, Netherlands.
4
Department of Medicine, Physiology, and Biophysics and Bioengineering, Division of Renal Diseases and Hypertension, University of
Colorado, Denver, CO and University of Colorado Hospital, 12605 E 16th Ave, Aurora, Colorado 80045, USA.
5
Center for Biomedical Research of the Canary Islands CIBICAN, University of La Laguna; Nephrology Service, Hospital Universitario de
Canarias, c/OFRA s/n, 38320, La Laguna, Tenerife, Spain.
6
Felsenstein Medical Research Center and Department of Nephrology, Rabin Medical Center, 39 Jabotinsky Street, 4941492, Petah
Tikva, Israel and Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
7
Department of Medicine, Complutense University, Instituto de Investigación, Hospital 12 de Octubre, Madrid, Spain and Department of
Nephrology, Hospital 12 de Octubre. Avda de Córdoba s/n. 28041 Madrid, Spain

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Individual podocytes fail and detach, causing localized Box 1 | BMI-based definitions of obesity
denudation of the glomerular basement membrane, sub-
sequent adhesions to the Bowman capsule and parietal • Normal weight: BMI 18.5–24.9 kg/m2
cell coverage, forming a nidus for the development of • Overweight: BMI 25–29.9 kg/m2
segmental sclerosis26,27. In the Fischer rat model, pro- • Obesity: BMI ≥30 kg/m2
teinuria and glomerulosclerosis were linearly related • Class 1 (or grade 1) obesity: BMI 30–34.9 kg/m2
to increasing body weight and could be accelerated • Class 2 (or grade 2) obesity: BMI 35–39.9 kg/m2
by unilateral nephrectomy, which promotes further • Class 3 (or grade 3) or morbid obesity: BMI ≥40 kg/m2
compensatory filtration demand25.
In addition to increased glomerular volume, ORG
biopsy samples show reduced glomerular density com- cellular stress35. Focal lipid vacuoles may also accumu-
pared to samples from non-obese kidney donors and late in the cytoplasm of mesangial cells and of tubular
patients with glomerulonephritis28. In so far as glo- epithelial cells, particularly in the proximal tubule8.
merular density can be used as a surrogate marker for A distinctive and diagnostically helpful feature of
nephron endowment, these data support the hypothesis ORG is the presence of relatively mild effacement
of low nephron endowment as a potential risk factor or of foot processes compared to primary FSGS. In a com-
priming event for the development of ORG. Autopsy parative study, mean foot process effacement was 40%
studies have shown that the number of nephrons at in ORG versus 75% in primary FSGS16. In some ORG
birth varies widely in children without known kid- biopsy samples, effacement can be as low as 25% despite
ney diseases29. The number of glomeruli per normal nephrotic-range proteinuria. In this respect, ORG
kidney ranges from approximately 225,000 to 1,825,000: resembles other adaptive forms of FSGS36, such as those
an eight‑fold difference30. Glomerular endowment that occur in patients with solitary kidney 37 or very low
strongly and inversely correlates with birth weight 31,32, birth weight 38. Foot process width is also greater in pri-
such that lower birth weight is associated with fewer mary FSGS than in adaptive forms, such as ORG39. This
nephrons and increased glomerular volume30. Precise difference likely reflects the underlying mechanisms of
quantification of nephron number using the ‘gold stand- podocyte injury. In primary FSGS, a presumably circu-
ard’ dissector/fractionator system at autopsy would, lating ‘permeability factor’ theoretically affects all podo-
however, be needed to differentiate to what extent the cytes uniformly, causing podocyte dysregulation and
reduced glomerular density seen in 2D histologic sec- toxicity, whereas in adaptive FSGS podocyte injury is
tions from patients with ORG reflects low nephron more heterogeneous and less severe, reflecting different
endowment versus tubular hypertrophy. stages of glomerular adaptation19,35. Animal models of
Approximately 50% of patients with ORG have mild ORG show predominantly intact foot processes with the
‘diabetoid’ changes despite a lack of clinically evident exception of podocyte denudation in foci of irreversible
glucose intolerance16. These changes include focal or podocyte stress25. Cell‑to‑cell propagation of podocyte
diffuse increases in mesangial matrix and thickening of injury contiguous with the initial foci of podocyte deple-
the glomerular basement membrane that is visible by tion might be mediated by interruption of pro-survival
light and electron microscopy, resembling mild diabetic signalling via nephrin or increased noxious stimuli,
glomerulosclerosis. In addition, focal intracellular lipid such as stretch tension and angiotensin II40,41. Spreading
vacuoles might accumulate in mesangial cells, podocytes fields of podocyte stress could promote enlargement of
and proximal tubular epithelial cells, as highlighted by segmental lesions, eventually resulting in global sclerosis.
Oil-Red‑O staining 33. These morphologic findings are
consistent with the transcriptional profiles of micro­ ORG superimposed on other renal disease
dissected ORG glomeruli, which indicate shared molec- Obesity is a risk factor for CKD and CKD progression in
ular pathways in ORG and diabetic glomerulosclerosis34. renal diseases other than ORG (BOX 2), and the develop­
Tubular atrophy and/or interstitial fibrosis (mean 1.26 ment of ORG does not preclude the development of
on a scale of 0–3+) and arteriosclerosis (mean 1.42 on other renal diseases. Thus ORG-associated glomerulo­
a scale of 0–3+) are common features, but the tubulo­ megaly and FSGS might occur superimposed on IgA
interstitial scarring and inflammation are generally less nephropathy or other renal conditions42. Moreover
severe than in primary FSGS and may be minimal16. obesity has been identified as an independent risk fac-
tor for end-stage renal disease (ESRD) in multivariable
Immunofluorescence and electron microscopy find- models after adjustment for multiple epidemiologic
ings. In ORG biopsy samples, immunofluorescence and clinical features including the presence of diabetes
reveals nonspecific trapping of IgM and complement mellitus and hypertension43. In a large historical cohort
C3 in lesions of sclerosis and hyalinosis; however, no study that used the Kaiser Permanente database, the
immune-complex-type deposits are seen. Podocytes adjusted relative risk for ESRD was 1.87 in overweight
overlying lesions of sclerosis may contain intracytoplas- individuals, 3.57 in those with class 1 obesity, 6.12 in
mic protein resorption droplets that stain for IgG, IgA those with class 2 obesity and 7.07 in those with class 3
and albumin. Using electron microscopy, corresponding obesity compared to individuals with normal BMI at
protein and lipid resorption droplets can be identified baseline43. These data support the hypothesis of obesity
focally within podocytes, where they might promote and probable ORG as potential cofactors in the progres-
endoplasmic reticulum (ER), oxidative, and autophagic sion of CKD of various aetiologies, and these trends

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 3


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

a b are borne out in clinical practice. Among 620 patients


with diabetes whose renal biopsy samples were ana-
lysed in 2011 at Columbia University, 33% had diabetic
nephropathy alone, 27% had diabetic nephropathy plus
a super­imposed nondiabetic renal disease, and 36% had
nondiabetic renal disease alone44. FSGS accounted for
22% of the cases of nondiabetic renal disease and 13%
of the cases of diabetic nephropathy plus nondiabetic
renal disease. In the majority (87%) of diabetic patients
with FSGS lesions, these lesions were attributed to ORG.

c d Subclinical renal changes


Systematic renal biopsies performed in morbidly obese
patients presenting for bariatric surgery have provided
a unique window onto early, subclinical renal changes.
In a Spanish study of 95 extremely obese patients
(mean BMI 53.6 kg/m2) with normal renal function, the
mean duration of obesity was 20 years, 39 (41%) patients
had microalbuminuria and four (4%) had albuminuria
(300–500 mg per day)45. Pathologic findings included
glomerulomegaly in 38% and FSGS lesions in only 5%
of patients. Using light microscopy, mesangial sclerosis
e f was identified in 60%, mild tubular atrophy and inter-
stitial fibrosis in 11% and arteriolosclerosis in 40% of the
pre-surgical biopsy samples45. An electron microscopy
study reported mild mesangial sclerosis, sparse parame-
sangial hyaline densities, glomerular basement mem-
brane thickening and variable podocyte effacement in the
majority of protocol biopsy samples from 13 morbidly
obese (BMI >50 mg/m2) patients without proteinuria who
were undergoing Roux‑en‑Y bypass surgery 46. Together,
these findings support the hypothesis that clinically
overt ORG is the ‘tip of the iceberg’ and that mild renal
g h pathologic alterations are already established in many
patients with morbid obesity and no clinically evident
renal disease47. Longer prospective follow‑up is needed
to determine whether patients with microalbuminuria
are at increased risk of developing ORG, analogous to the
greater risk of diabetic nephropathy that microalbum­
inuria confers among diabetic patients. As protocol renal
biopsies have been performed only in extremely obese
patients, little is known about the potential for subclinical
renal disease in those with class 1 or class 2 obesity.
Nature Reviews (ORG).
Figure 1 | Pathologic features of obesity-related glomerulopathy | Nephrology
The Clinical features of ORG
glomeruli of patients with ORG a | are hypertrophied in comparison to b | glomeruli
Incidence and prevalence
from non-obese age-matched control individuals (Jones methenamine silver stain,
×400). c | In patients with ORG, lesions of segmental sclerosis (arrow) tend to form The true incidence of ORG is unknown because renal
at the vascular pole (perihilar) and segmentally obliterate the capillary lumina biopsy policy in obese patients with proteinuria varies
(Periodic acid–Schiff stain, ×400). d | Focal dilatation of the afferent arteriole and widely between countries and centres, ORG can be pres-
the glomerular perihilar capillaries provides morphologic evidence for a high ent without clinical renal manifestations45, and in obese
single-nephron filtration rate (Periodic acid–Schiff stain, ×400). e | Some patients patients with type 2 diabetes mellitus (T2DM), it is often
with ORG show a mild increase in mesangial matrix and thickening of glomerular difficult to determine whether diabetes or obesity has a
basement membranes consistent with ‘diabetoid’ features (Jones methenamine predominant role in the development of proteinuria. In
silver stain, ×400). f | Glomerulus from a patient with advanced ORG showing a the absence of histological confirmation, the presence of
large segmental lesion of sclerosis (arrow) involving nearly half the tuft with persistent proteinuria in obese patients is generally con-
prominent tubular atrophy, interstitial fibrosis and chronic inflammation (Masson
sidered to be a surrogate marker of ORG, provided that
trichrome stain, ×200). g | Electron micrograph (×6,000) showing very mild
(approximately 15%) foot process effacement in a patient with ORG who had urine other renal diseases are not suspected. Observational
protein of 2.8 g per day. The podocyte cell body appears hypertrophied. h | The and epidemiological studies have reported that pro-
mesangial cells contain focal intracytoplasmic lipid vacuoles (arrows). A mild teinuria (semiquantative grade 1+ or greater) or macro­
increase in mesangial matrix and moderate foot process effacement are also albuminuria (albumin: creatinine ratio >300 mg/g) are
visible (electron micrograph, ×4000). prevalent in 4–10% of obese patients48–50.

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Presentation Box 2 | Obesity as a risk factor for renal diseases


Isolated proteinuria with or without renal impair-
ment is the most characteristic clinical presentation of Obesity is a significant risk factor for the progression of
ORG15–17,51,52. Other common findings are hypertension renal diseases other than ORG, particularly diabetic
(50–75% of patients) and dyslipidaemia (70–80% of nephropathy and IgA nephropathy. For example, in IgA
nephropathy, obesity increases the risk of hypertension,
patients)15–17,51,52. Subnephrotic proteinuria is found in
proteinuria, severe histologic lesions and end-stage renal
the majority of patients with ORG, particularly in stud- disease, and weight loss can result in reductions in
ies performed in China and Japan15,51,52. The reported proteinuria305.
percentage of patients with nephrotic-range protein-
uria ranges from 10% to 48%, but the presence of full
nephrotic syndrome is distinctly unusual (0–6%), even of glomeruli showing FSGS and global glomeruloscle-
in patients with massive proteinuria. The reasons why rosis as well as with the severity of tubulo­interstitial
patients with ORG do not develop the typical findings fibrosis and tubular atrophy 16,17,51,57. Correlations have
of nephrotic syndrome (hyperlipidaemia, hypoalbu- also been found between BMI and the extent of mesan-
minaemia and oedema) probably relate to the type of gial proliferation and podocyte hypertrophy in obese
podocyte injury and the fairly slow development of patients without overt renal disease45, and between
proteinuria in adaptive forms of FSGS16,17,53. The rarity reduced podocyte number (related in turn to the degree
of full nephrotic syndrome is a characteristic that ORG of glomerulomegaly) and proteinuria in patients with
shares with other entities in which glomerular hyper- ORG22. Low calorie diet-induced weight loss and other
filtration has a predominant pathogenic role, such as therapeutic interventions ameliorated renal histological
reflux nephropathy and FSGS secondary to reduced lesions in animal models of obesity 58,59, but no infor-
renal mass53–55. Some studies have suggested that the mation is available on the effect of weight loss on his-
tubular handling of proteins also might differ in these tological changes in patients with ORG. A single case
hyperfiltering nephropathies15,53. report of repeat biopsy in an obese child suggests that
The absence of full nephrotic syndrome despite sub- glomerulomegaly might be at least partially reversible
stantial proteinuria in patients with ORG has important following diet-induced weight loss60.
clinical implications; a progressive increase in protein-
uria can go undetected for years, leading to late clinical Predisposing factors
recognition. On the other hand, this peculiar character- A critical reduction in the number of functioning
istic of ORG is very useful diagnostically; obese patients nephrons is the central pathogenic factor in hyperfil-
may suffer diverse glomerular diseases16 and the pres- tering nephropathies. Although some patients with
ence of full nephrotic syndrome helps to distinguish substantial reductions in renal mass (owing to reflux
ORG from primary FSGS16,17,54,56 (TABLE 1). nephropathy, unilateral renal agenesis or extensive sur-
gical removal of renal parenchyma) develop proteinuria,
Disease course FSGS lesions and ESRD61, others maintain lifelong nor-
Few studies have examined the long-term outcomes of mal renal function without proteinuria. As a close linear
patients with ORG16,17,51. In the absence of therapeutic correlation between BMI and glomerular hyperfiltration
intervention, the clinical course is characterized by stable exists62, obesity could theoretically have a synergistic,
or slowly progressive proteinuria. Despite this indolent detrimental role in patients with reduced renal mass.
evolution, a substantial number of patients (10–33%) Supporting this hypothesis, in a series of patients who
may develop progressive renal failure and ESRD; had undergone unilateral nephrectomy, the only statis-
this percentage seems to increase with longer follow‑ tically significant difference between those who later
up16,17,51. Factors that are associated with progression of developed proteinuria and progressive renal impairment
renal failure include age and levels of serum creatinine and those who maintained normal renal function with
and proteinuria at presentation as well as time-averaged no proteinuria was a higher mean BMI in the former
proteinuria during follow‑up16,17,51. Comparative studies group, both at the time of nephrectomy and during
have shown a more sudden and aggressive disease pres- follow‑up63. In a retrospective study of patients with
entation in patients with primary FSGS than in those unilateral renal agenesis or a severe reduction of renal
with ORG as well as significantly lower renal survival mass as a result of medical or surgical causes, BMI at
in the former group (50% at 5 years and 25% at 10 years presentation and treatment with angiotensin-converting
versus 75% at 5 years and 50% at 10 years)16,17. enzyme (ACE) inhibitors were the only factors that were
associated with the development of proteinuria and loss
Clinical–pathologic correlations of renal function64.
A correlation between the percentage of glomeruli with Numerous experimental, clinical and epidemiologi-
FSGS and the amount of proteinuria or albuminuria has cal studies have shown that low birth weight, prematu-
been reported in patients with ORG17,45. However, ORG rity and intrauterine growth restriction are significantly
lesions can be found in patients without clinical evidence associated with increased risks of hypertension, cardio-
of renal disease45, and some patients with glomerulomeg- vascular events, T2DM, albuminuria and kidney disease
aly as the only histological manifestation of ORG have later in life; low nephron endowment is likely one of the
substantial proteinuria16. Renal functional impairment main pathogenic mechanisms that underlies this predis-
and progression to ESRD correlate with the percentage position31,65–69. Obesity might have a synergistic role in

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 5


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 1 | Clinical and pathologic differences between ORG-related and primary FSGS
Characteristic ORG-related FSGS Primary FSGS
Age at presentation Most common in middle-aged adults Most common in children and young adults
(mean age 37–46 years15–17,51,52), but may be
present in children and older adults
Clinical presentation Slowly progressive proteinuria Sudden onset of proteinuria with full
nephrotic syndrome in most cases
Proteinuria and serum • Sub-nephrotic proteinuria in 52–90% of • Nephrotic-range proteinuria in most
albumin patients patients
• Normal serum albumin levels • Hypoalbuminaemia is common
Full nephrotic syndrome* Unusual (<5% of patients) even in patients Common
with massive proteinuria
Clinical course • Slower progression than primary FSGS • Faster progression than ORG-related FSGS
• Renal survival 75% at 5 years and 50% at • Renal survival 50% at 5 years and 25% at
10 years16,17 10 years16,17
Glomeruli with FSGS Fewer than in primary FSGS (mean 12% of More frequent than in ORG-related FSGS
lesions glomeruli16) (mean 39% of glomeruli16)
FSGS variants Perihilar variant more common Not otherwise specified, tip and collapsing
variants more common
Glomerulomegaly Defining feature of ORG (100% of cases16) Variable (10% of cases16)
Foot process effacement Usually <50% glomerular surface area Usually >50% glomerular surface area
*Hypoalbuminaemia, hyperlipidaemia and oedema. ESRD, end-stage renal disease; FSGS, focal segmental glomerulosclerosis;
ORG, obesity-related glomerulopathy.

the development of renal complications in patients with than in those who were overweight or had normal BMI
a reduced nephron endowment. A retrospective study in (reductions of 86%, 45% and 42%, respectively). These
children with proteinuric kidney diseases showed that data suggest that obese patients might be more sensitive
both obesity and preterm birth significantly increased to the renoprotective effect of RAAS blockade than non-
the risk and rate of progression of renal disease70. Further obese patients. In some retrospective studies with longer
aggravating the problem, epidemiological studies have follow‑up, however, the beneficial effects of RAAS block-
shown that low birth weight is associated with increased ers became exhausted over time, partly due to further
weight gain during late childhood and adolescence, and weight gain in some participants17,51.
that individuals who experience cardiovascular events Accumulating data suggest that mineralocorticoid
often have a profile of low birth weight and subsequent receptor activation has a key role in obesity and the
obesity 71,72. The combination of low birth weight, child- metabolic syndrome75. Although very few studies have
hood malnutrition and adult obesity is particularly prev- addressed the effect of mineralocorticoid-receptor
alent in developing countries and has been proposed as blockers on the renal complications of obesity, a short-
one of the main causes of the increased prevalence of term study showed that the addition of spironolactone
CKD in these areas73. to an ACE inhibitor reduced albuminuria and blood
pressure in obese patients76.
Treatment of ORG
As in other chronic proteinuric nephropathies, a sig- Weight loss
nificant reduction in proteinuria is assumed to have a Non-surgical interventions. The effect of diet-induced
renoprotective effect in ORG. Renin–angiotensin–aldos- weight loss in obese patients with proteinuria has been
terone system (RAAS) blockade and weight reduction, evaluated in nonrandomized prospective studies57,77,78,
either diet-induced or promoted by bariatric surgery, are in randomized controlled trials (RCTs) that compared
the most studied therapeutic antiproteinuric measures. low calorie versus standard diets79–81, and in systematic
reviews and meta-analyses82–84. Some of these studies
RAAS blockade included obese patients with T2DM77–80, and in some
In retrospective studies, treatment of obese patients with studies hypocaloric diets were accompanied by pro-
proteinuria or biopsy-proven ORG using ACE inhibi- grammed exercise. Mean BMI ranged from 30–38 kg/m2
tors or angiotensin-receptor blockers (ARBs) induced at study entry and decreased significantly during follow‑
a substantial decrease in proteinuria to 30–80% of base- up in most studies, although there were notable differ-
line values16,17,51,53,54,64. Moreover, a post hoc analysis of ences between studies and patients depending on the
the REIN trial showed that the antiproteinuric effect type of diet, study duration and patient compliance.
of ramipril was significantly greater in obese and over- The most important overall finding was that weight
weight patients than in those with normal BMI74. The loss induced a significant decrease in proteinuria — the
reduction in incidence of ESRD with ramipril compared greater the weight loss, the greater the decrease. Notably, a
to placebo was also significantly greater in obese patients reduction in proteinuria was evident after only a few days

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

or weeks of a low-calorie diet and proteinuria decreased Dramatic favourable effects of bariatric surgery in
to remarkably low levels in those patients who experi- patients with massive proteinuria and ORG have been
enced the greatest weight losses. For instance, a RCT described in clinical case reports98,99. Bariatric surgery
showed a mean weight loss of 4% and a mean protein- may, however, be followed by severe renal complications,
uria decrease of 30% after 5 months of hypocaloric diet, such as nephrolithiasis, oxalate nephropathy and acute
but those patients who achieved weight losses of >6–10% kidney injury 93,100,101. Retrospective studies have shown
showed reductions in proteinuria to >60–70% of baseline an association between the number of postoperative
values80. A meta-analysis of nonsurgical weight-loss inter- complications and the presence of CKD101. Moreover, the
ventions in patients with CKD reported a mean reduction incidence of complications increased with CKD stage,
in BMI of 3.6 kg/m2 and a mean reduction in proteinuria ranging from 4.6% in those with normal renal func-
of 1.3 g per day during a mean follow‑up of 7.4 months82. tion or stage 1 CKD to 9.9% in those with stage 5 CKD.
Overall, no significant change in glomerular filtration Controlled studies of sufficient magnitude and duration
rate (GFR) was observed, although some RCTs showed in patients with substantial proteinuria, biopsy-proven
a trend towards a favourable effect of weight loss80,82. The ORG and CKD are needed to confirm the favourable
antiproteinuric effects of hypocaloric diets were accom- effects of bariatric surgery in these populations.
panied by substantial improvements in blood pressure,
lipid abnormalities, fasting glucose levels and insulin Pathogenesis of ORG
resistance57,77–84. However, the study durations were short Altered renal haemodynamics
(4 weeks to 2 years) and most included few patients. Renal plasma flow, glomerular filtration rate and filtra-
Two RCTs have compared the effects of weight loss tion fraction. The first cases of glomerular hyperfiltra-
(induced by low calorie diet or orlistat treatment) with tion in patients with ORG were reported 40 years ago14.
those of RAAS blockade in obese patients with protein- Since then, nine publications have reported GFR meas-
uria85,86. In these studies both interventions resulted in urements in obese and lean participants21,102–109. These
similar reductions in proteinuria. studies investigated renal haemodynamics in obese
individuals with preserved renal function; they were not
Bariatric surgery designed to investigate renal function in ORG. One study
Several RCTs have demonstrated favourable effects of bar- did not report data on proteinuria and albuminuria102 and
iatric surgery in obese patients with T2DM87,88. Moreover five studies included participants with microalbuminu-
the results of several uncontrolled studies suggest that ria21,103,104,107,108. As renal biopsies were not performed,
bariatric surgery might be beneficial in ORG82–84,89–96. the possibility cannot be excluded that some of the
These studies used various surgical procedures, microalbuminuric participants had ORG. In seven of
including Roux‑en‑Y gastric bypass, adjustable gastric the studies21,102,103,106–109, GFR uncorrected for body size
banding and sleeve gastrectomy. In comparison to the (BOX 3) was 12–61% higher in obese participants than in
participants of the dietary intervention studies discussed lean participants.
above57,77–84, patients who underwent bariatric surgery Among 11 studies21,103–112 that assessed renal plasma
had more severe obesity (BMI 44–53 kg/m2) and expe- flow (RPF) in obese and lean participants, eight stud-
rienced more dramatic weight loss (reduction in BMI ies showed a 9–33% increase in RPF21,103,106–109,111,112, two
of 11–21 kg/m2). A consistent finding was normaliza- showed no difference105,110, and one showed a decrease
tion of GFR after bariatric surgery in patients with glo- in RPF in those who were obese104. In three studies
merular hyperfiltration. Although the vast majority of filtration fraction (defined as GFR/RPF) was 9–29%
patients in the study cohorts had normoalbuminuria or higher in obese than in lean participants21,107,108, whereas
microalbuminuria, bariatric surgery reportedly induced a fourth study showed no difference in filtration fraction
consistent and significant decreases in albuminuria and between obese and lean groups103. Notably, two studies
proteinuria or even their resolution. These beneficial demonstrated that these renal haemodynamic changes
renal effects were accompanied by important improve- appear at an early stage of adiposity, when BMI is
ments in blood pressure and glycaemic control as well <30 kg/m2. The first of these studies showed that BMI
as favourable changes in metabolic and inflammatory is an independent predictor of filtration fraction and of
markers that persisted for 1–5 years after surgery. A RCT height-indexed GFR in a lean and overweight popula-
in obese patients with CKD confirmed these findings, tion62. The second reported that GFR, RPF and filtration
but the number of participants was very small (n = 11)97. fraction are ~11%, ~9% and ~4% higher, respectively, in
overweight than in lean particpants108.
The fact that RPF increases to a lesser extent than
Box 3 | Correction of GFR GFR in response to obesity implies the presence of renal
Glomerular filtration rate (GFR) measurements are commonly corrected for body surface vasodilation mainly or solely involving the afferent
area (BSA) in order to account for differences in body size. Indexing for BSA, however, arteriole. As the transcapillary hydraulic pressure dif-
leads to a systematic underestimation of GFR in patients with severe obesity that ference and ultrafiltration coefficient cannot be meas-
conceals glomerular hyperfiltration306. The main reason not to scale according to BSA is ured in humans, changes in these variables in obese
that the number of nephrons does not increase in parallel with adiposity. The increased patients have been estimated using measured GFR, RPF
GFR associated with obesity is the result of a raised single nephron GFR; non-corrected and the fractional urinary excretion of intravenously
GFR reflects this phenomenon, whereas it is obscured by indexing GFR for BSA. Thus, the administered neutral dextran21. Analysis of these data
use of BSA-indexed GFR should generally be abandoned in obese persons.
using a theoretical model of dextran transport through

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 7


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

a heteroporous membrane showed that an increase in RAAS overactivation might be involved in the patho-
transcapillary hydraulic pressure difference is the pre- genesis of hyperfiltration. Firstly, angiotensin II133 and
dominant factor that accounts for an increase in GFR in aldosterone134 vasoconstrict glomerular arterioles and
the setting of obesity 21. have a greater effect on the efferent than the afferent arte-
Systemic hypertension is highly prevalent in indi- rioles, which is expected to increase transcapillary hydrau-
viduals who are overweight or obese113. The findings of lic pressure difference and GFR133. Moreover aldosterone
renal vasodilation and increased transcapillary hydraulic has been shown to increase GFR in humans135. Secondly,
pressure difference21 in obese patients suggest that sys- angiotensin II increases sodium reabsorption proximally,
temic hypertension might contribute to the pathogenesis by stimulation of the luminal Na+-H+ exchanger and the
of hyperfiltration in these patients as an abnormal trans- basolateral Na+-K+-ATPase, and distally, by activation
mission of increased arterial pressure to the glomerular of the epithelial Na+ channel (ENaC)136. Angiotensin II
capillaries through a dilated afferent arteriole is expected might also directly activate the mineralocorticoid recep-
to augment transcapillary hydraulic pressure difference tor 137, resulting in increased sodium reabsorption and
and thus increase GFR21. a positive sodium balance. RAAS overactivation might
A small study assessed renal haemodynamics in therefore lead to excessive sodium reabsorption and
eight patients with severe obesity before and 1 year after resulting hypertension and hyperfiltration138,139.
bariatric surgery 89. Following surgery, the mean BMI of In obese individuals the renal sympathetic nervous
these patients decreased from 48 kg/m2 to 32 kg/m2, their system (RSNS) is also overactivated140,141 and induces
arterial pressure normalized and their GFR, RPF and sodium retention142. Three factors that are associated with
filtration fractions decreased by means of 24%, 13% obesity activate the RSNS: leptin, low adiponectin levels
and 11%, respectively. This investigation demonstrates that and obstructive sleep apnea (OSA). Circulating leptin
obesity-related glomerular hyperfiltration is reversible levels are increased in obesity and activate the RSNS143,144,
following weight loss. which promotes sodium retention and hypertension145.
Consistent with this causal role, mice that were deficient
Tubular sodium reabsorption. In obese individuals in the leptin receptor did not develop hypertension when
the filtered sodium load is increased in proportion to they became obese146, and even severely obese patients
the degree of hyperfiltration; therefore, increased reab- with loss-of-function mutations in the leptin receptor
sorption of salt along the nephron is indispensable to pre- have low blood pressure147. Low adiponectin levels in
vent volume depletion. In the setting of obesity; however, obese patients might also activate the RSNS and promote
salt is reabsorbed to excess. BMI and waist circumference salt retention148. Moreover OSA is frequently associated
are independent predictors of the proximal reabsorption with obesity and overactivates the RSNS. A study showed
of sodium114, and this reabsorption is increased in white that a group of patients with moderate to severe OSA
patients with obesity 107,115. Data from experimental stud- had normal GFRs with low RPFs149. Treatment with con-
ies suggest that increased sodium reabsorption in the tinuous positive airway pressure increased RPF, whereas
setting of obesity might result from increased activation GFR remained unchanged. The decreased RPF before
of sodium transporters along the nephron116,117. treatment is at variance with the more frequent vasodi-
lation pattern seen in severe obesity, probably reflecting
Glomerular hyperfiltration. Two concepts have been the very high degree of RSNS activity induced by OSA.
proposed to explain obesity-related glomerular abnor- Hyperinsulinaemia secondary to insulin resistance
malities (FIG. 2). The classic concept is the primary hae- probably also has a role in obesity-associated renal
modynamic hypothesis, in which the primary event is dysfunction. Although experimental studies suggest
vasodilation of the afferent arteriole, resulting in glomer- direct effects of insulin on the glomerular microcircu-
ular hyperfiltration118. The second proposed mechanism lation150–152, these effects do not fit with the glomerular
is the tubulocentric hypothesis119–121, in which the primary haemodynamics found in obese individuals. Insulin
event is increased proximal reabsorption of salt and water, increases the tubular reabsorption of sodium by stim-
resulting in decreased solute delivery to the macula densa, ulating the activity of ENaC in the late distal tubule153,
deactivation of tubuloglomerular feedback, preglomerular and less prominently in the proximal tubule154 and the
vasodilation and consequent glomerular hyperfiltration. loop of Henle155,156. Together these effects contribute to
Proximal sodium reabsorption is increased in the set- the pathogenesis of salt retention in obesity.
ting of obesity 107,114,115,122 and the carboanhydrase inhib-
itor acetazolamide was shown to decrease GFR by 21% Protein intake. A large population-based survey that
in nondiabetic, severely obese patients123. These effects used 24 h urine collections identified protein intake
are compatible with the tubulocentric theory, but do not as a predictor of GFR, independent of obesity and salt
prove or disprove either concept. intake157. Most experimental human and animal stud-
ies have shown that habitual protein intake modulates
Hormonal and neurohormonal activation. In the set- GFR158,159. A study in obese individuals reported that
ting of obesity the RAAS is overactivated and levels of switching from a low-protein to a high-protein diet
RAAS components are increased in the circulation124–129 induced only a <5% increase in GFR160, suggesting that
and in renal tissue126,130. This finding is explained in glomerular hyperfiltration is mostly determined by fac-
part by the capacity of fat cells to synthesize RAAS tors that are unrelated to protein intake, at least in the
components130–132. short-term.

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Systemic • High salt diet


hypertension • Low nephron
Systemic or local number
Deactivation of • High protein diet
tubuloglomerular intrarenal factor(s)
feedback
?
↓ Solute delivery Afferent ↑ Single nephron ± Efferent arteriole
to macula densa arteriole dilation plasma flow constriction

↑ Glomerular • Ang II
pressure • Aldosterone

↑ Filtrate flow (single ↑ Glomerular


nephron filtration rate) capillary wall
tension stress
Shear stress on podocytes
↑ Proximal tubular Basement
salt reabsorption membrane
Mechanotransduction:
↑ Ang II, AT1R, TGF-β and TGF-βR distension

• Ang II Podocyte hypertrophy Capillary growth and


• Renal sympathetic and apoptosis Mismatch glomerulomegaly
nervous system
• Insulin
• ↑ Peritubular Podocyte detachment
oncotic pressure
• Microvilli
mechanosensing
and transduction Focal segmental glomerulosclerosis—obesity-related glomerulopathy

Figure 2 | Haemodynamic alterations in obesity. Primary dilatation of the afferent arteriole and variable constriction of
Nature in
the efferent arteriole via activation of angiotensin II (Ang II) and aldosterone contribute to increases Reviews | Nephrology
single nephron
plasma flow, glomerular intracapillary hydrostatic pressure, and filtration rate. The major driver of afferent arteriolar
dilatation is unknown, but deactivation of tubuloglomerular feedback via increased proximal tubular salt reabsorption
and decreased delivery to the macula densa likely has a role. A host of factors, including Ang II, the renal sympathetic
nervous system, insulin, an increase in postglomerular oncotic pressure due to increased filtration fraction, and
mechanosensors of tubular flow rates, mediate the increased tubular reabsorption of sodium. The increase in filtrate
flow (single nephron filtration rate) in turn promotes glomerular capillary wall stretch tension, glomerulomegaly, and
maladaptive podocyte stress leading to obesity-related glomerulopathy and focal segmental glomerulosclerosis. AT1R,
type 1 angiotensin II receptor; TGF‑β, transforming growth factor β; TGF‑βR, TGF‑β receptor.

Physiologic consequences. The prevalence of albuminu- high filtration fractions had high proximal reabsorption
ria is increased in people who are obese161,162. Various of sodium107. Thus, glomerular hyperfiltration might
factors contribute to this increased albumin excretion: enhance sodium reabsorption by raising peritubular
increased capillary pressure and transcapillary hydraulic oncotic pressures.
pressure difference, increased wall tension, glomerular Increased GFR leads to increased proximal tubular
tuft enlargement and stretching of glomerular cells. In flow. The apical microvilli of the proximal tubular epi-
addition, angiotensin II might directly affect glomeru- thelia function as a flow sensor, leading to activation of
lar permselectivity 163 independently of filtration pres- sodium transporters and increased sodium reabsorp-
sure. Other factors, such as low circulating adiponectin, tion169. In this manner, increased axial luminal flow in
might induce albuminuria164,165 through a direct action obesity-induced hyperfiltration might directly modulate
on podocytes. proximal tubule sodium transporters.
Increased peritubular oncotic pressure also affects
salt handling. Obesity-associated hyperfiltration is Structural consequences. Increased hydrostatic pressure
usually associated with an increased filtration fraction. in the glomerular capillaries (glomerular hypertension)
The expected consequence is haemoconcentration in drives an increased circumferential and axial capillary
the postglomerular circulation and an increase in the wall stress, which induces basement membrane expan-
oncotic pressure of the plasma entering the peritubular sion and glomerulomegaly as well as an increase in the
capillaries. As one of the major determinants of proximal ultrafiltrate flow entering the Bowman space (hyperfil-
tubular reabsorption is the pressure gradient determined tration)170,171. Hyperfiltration exerts a high fluid shear
by Starling forces, this augmented oncotic pressure is stress on podocytes, prompting maladaptive hypertro-
expected to promote proximal tubular sodium reabsorp- phy, which ultimately leads to podocyte detachment and
tion166–168. In a study of obese individuals, those who had global glomerulosclerosis. Fluid shear stress activates
filtration fractions within the normal range had normal various mediators, such as angiotensin II, angiotensin II
proximal reabsorption of sodium, whereas those with type 1 receptor, transforming growth factor β (TGF-β),

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 9


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

TGF-β receptor and phospholipase D, which might be filtered by glomeruli and taken‑up by megalin to sup-
involved in podocyte hypertrophy, apoptosis, decreased port lysosomal functions of proximal tubular cells186.
adhesion and detachment. Obese adults with normo­ Whether uptake of adipose tissue-derived cathepsins by
albuminuria or mildly increased albuminuria excrete podocytes could induce maladaptive responses remains
increased levels of urine podocyte-associated mRNA, speculative. In podocytes, cytosolic cathepsin L has
reflecting early-stage podocyte injury 172. been shown to proteolyze dynamin, synaptopodin,
Data from animal and human studies suggest that the and CD2‑associated protein, thereby contributing to
areas of the glomerular and tubular spaces are increased secondary FSGS and proteinuria187–189. As cystatin C is
in the setting of obesity. In a dog model of obesity with the natural inhibitor of cathepsins and pharmaceutical
hyperfiltration, the area of the Bowman space was 41% companies have specific cathepsin inhibitors under clin-
greater than in lean dogs173. Moreover, an analysis of ical investigation, cathepsin and cystatin homeostasis in
biopsy samples from nondiabetic patients showed that obese or diabetic CKD is rapidly gaining interest.
the areas of the Bowman space and proximal tubular
lumen were 41% and 54% higher, respectively, in those Risks of different adipose compartments
from obese patients with high GFR, than in those from BMI differentiates poorly between body fat, muscle mass
lean patients with normal GFR174. Considering the high and water, and does not take into account the wide range
GFR of the obese cohort, it is reasonable to assume that of body fat distribution in lean and obese adults. Several
the glomerular and tubular urinary spaces expanded as a studies have shown that abdominal or central obesity,
consequence of the high ultrafiltrate flow, representing a reflected as waist circumference or waist‑to‑hip ratio
morphologic sign of glomerular hyperfiltration. (WHR), is a better marker of cardiometabolic risk and
mortality than is BMI and is more closely associated with
Maladaptive cell and hormone responses renal functional impairment 190,191. However, WHR and
Adipose tissue as an endocrine organ and potential waist circumference discriminate suboptimally between
renal effects. Adipose tissue is a pleiotropic source of abdominal subcutaneous (SAT) and visceral adipose
hormones and chemokines, collectively called adi- tissue (VAT), which can be assessed by CT or MRI. This
pokines175. In obesity, satiated fat cells facilitate adipose distinction is important because these compartments
tissue expansion by secreting angiogenic and inflam- convey different metabolic risk192. VAT demonstrates a
matory adipokines (such as angiopoietins, vascular larger adipokine response to nutrients than does SAT193.
endothelial growth factor (VEGF), cathepsins and cys- Although SAT is the major site of fat accumulation, VAT
tatin‑C) that promote stromal rearrangements, neovas- is a better predictor of T2DM and risk factors for cardio­
cularization and the formation of novel adipocytes176 vascular disease. As the risk of CKD increases with the
(FIG. 3). White adipocytes mature from pericytes of the number of cardiovascular risk factors194, VAT might be a
adipose microvasculature177. Angiogenesis and adipo- better predictor of CKD than are conventional measures
genesis are intricately linked, and disturbed angio­genesis of obesity. In the Framingham Offspring cohort, VAT
has been associated with diminished adipose tissue and SAT were not independently associated with CKD
expandability 178. The paracrine roles of adipokines in the when GFR was estimated using a creatinine-based equa-
adipose tissue might have distant effects in the kidney. tion195, but both VAT and SAT were associated with CKD
In experimental settings adipokines stimulate kidney cells when a cystatin C‑based equation was used196.
to undergo adaptive or maladaptive responses to Impaired adipose tissue expandability and an
cope with the mechanical forces of hyperfiltration179. increased sensitivity of VAT to lipolysis might lead to
Adipokines such as leptin, adiponectin, and resistin an overflow of nonesterified fatty acids (NEFA) into the
affect cellular hypertrophy, extracellular matrix and circulation, resulting in deposition in non-adipose tis-
renal fibrosis179, whereas factors such as angiopoietins sues such as liver, heart and kidney 197. The arche­typical
and VEGF maintain endothelial–pericyte integrity 180. example of ectopic lipid is non-alcoholic fatty liver
Mesangial cells are specialized pericytes that support disease (NAFLD), which might contribute to hepatic
glomerular capillaries and regulate capillary flow 181. insulin resistance, gluconeogenesis, and increased sys-
Experimental models of mesangiolysis and pericyte temic levels of inflammatory adipokines198, and thus
loss show glomerular microaneurysm formation and confer independent risk of CKD. A meta-analysis
capillary ballooning that might contribute to glomeru- showed that NAFLD was independently associated with
lomegaly 182. A mesangial imbalance in VEGF and angio- CKD, although none of the included studies adjusted
poietins was associated with disruption of the glomerular for VAT199.
capillary structure in vitro183. In pigs, diabetes and an BMI correlates with triglyceride accumulation in the
atherogenic diet caused systemic capillary tortuosity, human kidney cortex 33. In human nephrectomy speci-
and concomitantly, an imbalance in renal angiopoie- mens from obese individuals, lipid was predominantly
tins, which correlated with the development of diabetic found in the proximal tubules and to a lesser degree in
nephropathy 184. Another study showed a possible role of glomeruli33. Ectopic lipid in the kidney or a ‘fatty kidney’
cathepsin S in protease-activated receptor‑2‑mediated may constitute a quintessential biomarker of obesity-
endothelial dysfunction in human and experimental related kidney disease8. Ample experimental studies
diabetic nephropathy 185, raising the question of whether underscore the role of lipotoxicity or lipid overload in
circulating cathepsin S from adipose tissue could con- obesity-related kidney disease, but human translation
tribute to this effect. Circulating cathepsin B may be is hampered by the lack of a non-invasive method to

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Microcirculation Adipokines Normal Obesity

Afferent Mesangial
NEFA cell
arteriole
Mesangial
Matrix: matrix
• Cathepsins
• Cystatin
• MMPs

Angiogenesis: Glycocalyx
Angiogenesis • VEGFs ↑ Radius
• Angiopoietins ↑ Tension
Podocyte Pressure
Pericyte
Metabolism:
• Aldosterone
• Ang II
Adipose tissue expandability • Leptin
• Adiponectin

Inflammation:
• TGF-β Endothelium
• IL-6 or IL-1
• TNF Albumin
• MCP-1 Efferent
Lipid droplet arteriole
Hypoxia:
HIF-1
Macrophage Adipocyte Tubular cell

Figure 3 | Potential role of adipokines in the development of obesity-related glomerulopathy.NatureInReviews


obesity,|satiated fat
Nephrology
cells facilitate adipose tissue expansion by secreting angiogenic and inflammatory adipokines such as angiopoietins,
vascular endothelial growth factor (VEGF), and cathepsins. These adipokines promote stromal rearrangements,
neovascularization and the formation of novel adipocytes from pericytes of the microvasculature. Angiogenesis and
adipogenesis are intricately linked, and disturbed angiogenesis has been associated with diminished adipose tissue
expandability. Systemically circulating adipokines might reach the kidney and have local effects on mesangial cells,
podocytes, and tubular cells, promoting maladaptive responses to glomerular hyperfiltration and albuminuria. Ang II,
angiotensin II, HIF‑1, hypoxia-inducible factor‑1; MCP‑1, monocyte chemoattractant protein‑1 (also known as C‑C motif
chemokine 2); MMP, matrix metalloproteinase; NEFA, non-esterified fatty acid; TGF‑β, transforming growth factor β.

assess the fatty kidney. Current developments in clinical and podocyte ANGTPL4 induced proteinuria in rats207.
proton magnetic resonance spectroscopy of the kidney These findings provide a likely molecular underpinning
show promise200,201 but await validation in interventional of the Moorhead hypothesis; concomitant hyperlipi-
studies and against human biopsy samples. daemia and albuminuria cause self-perpetuating renal
disease through the accumulation of lipid in the injured
Effects of ectopic lipid on glomerular integrity, albu- kidney leading to glomerulosclerosis208.
minuria and fibrosis. Mesangial cells accumulate lipids Glomerulosclerotic pathways have also been linked
via various receptors and may transform into a type of to fatty acid metabolism. Palmitate was found to upregu-
foam cell8. Mesangial lipid accumulation via insulin late Smad3 in mice, resulting in loss of synaptopodin and
growth factor‑1 has been associated with loss of con- mitochondrial injury in podocytes209. Moreover Smad3
tractile function in vitro and might contribute to loss deficiency protected mice from obesity-induced podo-
of glomerular integrity 202,203. Moreover inflammation cyte injury that otherwise resulted in markedly increased
interferes with LDL-receptor feedback regulation in mitochondrial biogenesis and respiration before the
mesangial cells leading to unopposed lipid accumula- onset of systemic and renal insulin resistance. TGF‑β/
tion204. In an elegant porcine study that used micro‑CT, Smad3 signalling constitutes a major fibrotic pathway,
increased renal triglyceride accumulation was associated but is also involved in regulating insulin gene transcrip-
with microvascular proliferation and increases in GFR, tion210 and adipose tissue homeostasis211. In mice, Smad3
expression of pro-angiogenic and inflammatory fac- inhibition caused a phenotypic change of adipocytes
tors and albumin leakage205. Plasma albumin normally (from white to brown adipose tissue) that was associ-
carries >99% of circulating NEFA and albumin-bound ated with increased PPARγ co-activator‑1α (PGC1α)
NEFA has been shown to induce massive macropino­ expression211. This fibrotic pathway is thought to impair
cytosis in podocytes206. This finding suggests that podo- fatty acid oxidation in renal epithelial cells by repressing
cytes sense the disruption of glomerular integrity via PGC1α‑induced expression of rate-limiting enzymes
NEFA. In pronounced albuminuria, the serum ratio for β‑oxidation such as carnitine palmitoyltransferase‑1
of free NEFA to albumin-bound NEFA and the plasma (CPT1)212.
levels of angiopoietin-related protein 4 (ANGPTL4) In nephrotic-range proteinuria, NEFA-bound albu-
are increased207. ANGPTL4 aggravates hypertriglyceri- min is taken up by proximal tubular cells leading to giant
demia by inhibiting lipoprotein lipase. NEFA have been lipid droplets. Tubular uptake of NEFA occurs propor-
shown to induce ANGPTL4 expression in podocytes206 tionally to plasma NEFA concentration (basolateral side)

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 11


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

and filtered albuminuria (luminal side)213. NEFA are diabetes217. Insulin resistance has been independently
the predominant fuel for the highly energetic tubular associated with renal hyperfiltration and incident CKD
transport processes, but overabundant NEFA are incor- in large cohort studies194,218,219. The effects of insulin on
porated into lipid droplets as triacylglycerol214. This the human kidney are complex and include both pressor
abundance might interfere with normal mitochon- and depressor effects, but the generally accepted view
drial function depending on the fatty acid oxidation is that short-term insulin infusion increases GFR and
capacity of the cell215. Impaired fatty acid oxidation has distal tubular sodium reabsorption220.
been shown to cause lipid accumulation and has a key Podocytes express insulin receptors and, via insulin
role in the development of renal interstitial fibrosis212. signalling, potentially adjust their morphology to post-
Consistent with this finding, a genome-wide transcrip- prandial changes in intracapillary pressure and GFR221.
tome analysis of renal tubule samples identified dysreg- Accumulation of NEFA in podocytes has been linked to
ulation of inflammatory and metabolic pathways as well insulin resistance222 and podocyte apoptosis in vitro223.
as reduced levels of the rate-limiting enzymes of fatty Normal insulin/phosphatidylinositol 3‑kinase (PI3K)/
acid oxidation, CPT1 and PGC1α, in patients with CKD Akt and mTOR signalling (FIG. 4) seem to be critical for
plus hypertension and/or diabetes212. podocyte function and survival and mouse models with
disturbances in these pathways show ORG-like fea-
Insulin resistance and mTOR signalling. Levels of tures25,224,225. Insulin also stimulates VEGF production
ectopic lipids correlate more strongly with insulin resist- in podocytes226. Mice with podocyte-specific knockout
ance than with any of the commonly used indices of of the insulin receptor developed the histological fea-
obesity such as BMI, waist circumference and WHR216. tures of ORG, but maintained normal glucose levels224.
Cellular insulin resistance was originally proposed by A contrary characteristic was the absence of the renal
Reaven as a unifying concept of unhealthy obesity, enlargement and cellular hypertrophy seen in patients
which is the clustering of obesity with cardiovascular with ORG. A possible explanation is that absent insu-
risk factors such as dyslipidaemia, hypertension and lin signalling impaired mTOR activation. An elegant

• Growth factors
Insulin receptor • Nutrients

IRS-1
mTORC2

PI3K Akt

SGK1 Akt • PKCα


• Rho or Rac
TSC1/2
Sodium FoxO
reabsorption Actin
Rheb Rheb
(activation remodelling
GDP GTP of ENaC in Cell
tubular cells) survival

mTORC1

S6K
• Protein synthesis
• Cellular growth
4E-BP1
• SREBP • HIF-1
• PPARγ • VEGF hATG1

• Adipogenesis Angiogenesis Autophagy


• Lipid synthesis
Figure 4 | The insulin/ PI3K/Akt/ mTOR pathway. The mTOR protein kinase complexes mTORC1 and mTORC2
constitute a sensor that integrates various metabolic and hormonal cues to regulate cellular growth and homeostasis.
Nature Reviews | Nephrology
Insulin signalling via phosphatidylinositol 3‑kinase (PI3K) can stimulate mTORC1 and the downstream pathways of
lipogenesis, angiogenesis and cellular growth (protein synthesis) as well as a decrease in autophagy. The downstream
target of mTORC1, S6 kinase (S6K), negatively regulates insulin receptor substrate 1 (IRS‑1), causing resistance to insulin
signalling. Nutrients and growth factors may activate mTORC2, leading to actin remodelling, changes in cell phenotype
and survival, and sodium reabsorption via activation of the epithelial Na+ channel (ENaC) in tubular cells. Activation of
mTORC2 might also lead to insulin resistance via inhibition of IRS‑1. 4E‑BP1, 4E‑binding protein 1; FoxO, Forkhead box
protein O1; GDP, guanosine diphosphate; GTP, guanosine‑5ʹ‑triphosphate; hATG1, autophagy-related protein 1
homologue; HIF‑1, hypoxia-inducible factor‑1; PKCα, protein kinase C α type; PPARγ, peroxisome proliferator-activated
receptor γ; Rheb, GTP-binding protein Rheb; SGK1, serine/threonine-protein kinase Sgk1; SREBP, sterol regulatory
element-binding protein; TSC, tuberous sclerosis protein; VEGF, vascular endothelial growth factor.

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

study in a transgenic rat model showed that podocytes Insulin decreases gluconeogenesis in the kidney similar to
undergo hypertrophy in a mTOR-dependent manner its effects in the liver 231. Lipid accumulation in proximal
to cover the enlarging glomerular tuft in response to tubular cells could drive renal gluconeogenesis through
increasing body weight 25. Failure of podocytes to hyper- an increase in acetyl-coenzyme A concentration232. In
trophy via mTOR in response to glomerulomegaly led T2DM, the kidney is thought to contribute to hyper­
to focal denudation of the glomerular tuft and an ORG- glycaemia233,234. Obesity and T2DM do not seem to inter-
like phenotype. Similarly, Akt2 signalling (activated by fere with tubular insulin signalling in rodent models235,
mTORC2) was essential for podocyte compensation suggesting continued adaptation to metabolic load.
after nephron reduction in a mouse model and for
podocyte survival in human CKD225. The mTOR com- Abnormal lipid metabolism
plexes (mTORC1 and mTORC2) constitute a sensor that In ORG, abnormal lipid metabolism promotes increased
integrates different metabolic and hormonal signals to triglyceride and cholesterol ester accumulation in the
regulate glomerular homeostasis and adaptive growth227. kidney 5–8. Several proteins with roles in these processes
Insulin signalling can stimulate mTORC1 and down- have shown promise as potential therapeutic targets in
stream pathways of lipogenesis (via PPARγ and sterol animal models of obesity.
regulatory element-binding protein‑1 (SREBP‑1)),
angiogenesis (via VEGF), and cellular growth (via S6 Fatty acid and triglycerides. Increased triglyceride
kinase and 4E‑binding protein 1), whereas stimulation accumulation can occur as a result of increased fatty
of mTORC2 can lead to actin remodelling, cell survival acid synthesis, increased uptake via CD36, or decreased
(via forkhead box protein O1) and sodium reabsorption fatty acid oxidation (FIG. 5). SREBP‑1 is a master regula-
(via serine/threonine-protein kinase Sgk1). Diabetic tor of fatty acid and triglyceride synthesis236–243. In the
nephropathy has been associated with increased mTOR murine high fat diet (HFD)-induced obesity model (see
signalling 228,229 but the contribution of this signalling Supplementary information S1 (box)), SREBP‑1 expres-
pathway has not been evaluated in ORG. sion and activity results in increased renal lipid accumu-
Insulin signalling also has an important role in prox- lation244–256. Studies using SREBP‑1a transgenic mice have
imal tubular cells. A mouse model with knockout of the provided evidence for a direct role of SREBP‑1 in mediat-
proximal tubule insulin receptor showed hyperglycaemia, ing renal lipid accumulation and renal disease; increased
apparently as a result of increased renal gluconeogenesis230. renal expression of SREBP‑1a resulted in proteinuria,
mesangial expansion and increases in inflammation,
oxidative stress and expression of profibrotic growth
FXR ↑ CD36 and ↑ FATP FXR factors244. The effects of HFD and obesity on kidney
disease are prevented in SREBP‑1c knockout mice247.
↑ SREBP-1c ↓ PPARα Increased SREBP‑1 activation induces expression of
↑ Fatty acid uptake
proinflammatory cytokines, several of which in turn acti-
↑ ACC, ↑ FAS ↓ ACOX1 vate SREBP‑1 (including tumour necrosis factor (TNF),
and ↑ SCD-1 and ↓ CPT
IL‑1β, and interferon‑γ)245, resulting in a potentially
↑ Fatty acid ↓ Fatty acid vicious cycle to accelerate lipid accumulation and inflam-
↑ Triglycerides
synthesis oxidation mation. In addition to high glucose, saturated fatty acids
↓ MCAD and oxidized phospholipids, several vasoactive hormones
↑ LPK and ↓ LCAD and profibrotic growth factors, including angiotensin II,
↑ Lipid
↓ Triglyceride induce upregulation of SREBP‑1 (REFS 250,257), which in
droplet
hydrolysis
↑ ChREBP proteins ↓ SIRT3 turn mediates TGF‑β signalling 258,259.
Cell culture experiments have provided insights into
FXR ↓ Lipolysis TGR5 the mechanisms that regulate high-glucose-induced
increases in SREBP‑1 activation. Signalling through
Figure 5 | Fatty acid and triglyceride metabolism in obesity-related glomerulopathy.
PI3K, Akt, mTOR, glycogen synthase kinase‑3β, JAK2/
Renal triglyceride accumulation can occur as a result of increased
Nature fatty acid| synthesis
Reviews Nephrology
mediated by sterol regulatory element-binding protein 1c (SREBP‑1c) and its target STAT1, and high mobility group protein B1 upregulate
enzymes including acetyl-CoA carboxylase (ACC), fatty acid synthase (FAS), and the expression of SREBP‑1, whereas PTEN prevents
stearoyl CoA desaturase‑1 (SCD‑1); and/or mediated by carbohydrate-responsive SREBP‑1 expression260–266.
element-binding protein (ChREBP) and its target enzymes including liver pyruvate kinase The importance of SREBP‑1 in mediating lipid accu-
(LPK). Renal triglyceride accumulation can also occur by increased uptake via CD36 or mulation and increased expression of proinflammatory
fatty acid transport protein (FATP); or by decreased fatty acid oxidation mediated by cytokines and profibrotic growth factors, as well as evi-
peroxisome proliferator-activated receptor α (PPARα) and its target enzymes, or dence for increased SREBP‑1 expression in the glomeruli
NAD-dependent protein deacetylase sirtuin‑3 (SIRT3) and its target enzymes. TGR5 of patients with ORG34, have led to a search for in vivo
agonists decrease inflammation and mitochondrial ROS generation and increase inhibitors of this protein. The farnesoid X‑activated
mitochondrial biogenesis, mitochondrial antioxidant generation and mitochondrial fatty
receptor (FXR) agonists inhibit SREBP‑1 expression, lipid
acid β‑oxidation. As well as decreasing inflammation, oxidative stress and fibrosis, FXR
agonists decrease lipid accumulation by inhibiting SREBP‑1 and ChREBP and accumulation, inflammation and fibrosis in animal mod-
stimulating PPARα. ACOX1, peroxisomal acyl-coenzyme A oxidase 1; CPT, carnitine els of diet-induced obesity and insulin resistance267–270.
O‑palmitoyltransferase; FXR, farnesoid X‑activated receptor; TGR5, G‑protein coupled In addition, vitamin  D receptor (VDR) agonists
bile acid receptor 1; LCAD, long-chain specific acyl-CoA dehydrogenase, mitochondrial; inhibit SREBP‑1, lipid accumulation, inflammation
MCAD; medium-chain specific acyl-CoA dehydrogenase, mitochondrial. and fibrosis271.

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 13


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

An alternative approach to limit fatty acid and triglyc- mechanisms include roles for inflammatory cytokines,
eride accumulation is to increase fatty acid oxidation via which interfere with the SCAP–SREBP‑2–LDL receptor
activation of PPARα. This approach is relevant because and 3‑hydroxy‑3‑methylglutaryl-CoA reductase path-
kidney biopsy samples from patients with ORG and ways203,285, the presence of ER stress, which stimulates
diabetes show decreased expression of PPARα and tar- SREBP‑2 release from the ER286, and advanced glycation
get enzymes that mediate fatty acid oxidation212,272. The end products, which increase abnormal translocation of
PPARα agonist fenofibrate prevented development of kid- SCAP from the ER to the Golgi resulting in activation
ney disease in HFD-fed mice273, in diabetic db/db mice274, of SREBP‑2 (REF. 287).
and in Zucker diabetic fatty rats275. In addition, several Small molecule inhibitors of SREBP‑2 are the subject
large-scale studies276 including DAIS, FIELD, the FIELD of ongoing research. Studies have shown, however, that
washout sub-study, and the ACCORD trial showed that caloric restriction in ageing 249 and treatment of mice
fibrates reduce progression of albuminuria, and despite with HFD-induced obesity using a vitamin D receptor
an initial transient reduction in GFR, these agents might agonist 271 decrease SREBP‑2 expression and activity
reduce loss of renal function in the long term277–281. in the kidney, prevent cholesterol accumulation and
Direct activation of AMPK with AICAR or treat- improve renal disease. Moreover, angiotensin 1–7 has
ment with metformin also increases fatty acid oxidation been shown to decrease renal inflammation and renal
and prevents the development of HFD-induced kid- injury in HFD-fed mice by downregulating the SCAP–
ney injury 282,283. Treatment with a selective G‑protein SREBP‑2–LDL receptor pathway 288,289. Statin treatment
coupled bile acid receptor 1 (TGR5) agonist increased of HFD-fed mice has also been shown to reduce lipid
mitochondrial fatty acid β‑oxidation and prevented lipid accumulation in the proximal tubules, reduce glomeru-
accumulation and the development of renal disease in lar hypertrophy, increase podocyte nephrin expression
mice with HFD-induced obesity and in db/db mice284. and decrease desmin expression290.
An alternative approach to limit cholesterol accumu-
Cholesterol. Increased cholesterol accumulation can lation is to increase cholesterol efflux. The nuclear recep-
occur as a result of increased cholesterol synthesis, tor LXR has a major role in cholesterol efflux, decreases
increased cholesterol uptake or decreased cholesterol inflammation and prevents the development of ather-
efflux (FIG. 6). SREBP‑2 is a master regulator of choles- osclerosis291–293. LXR also induces cholesterol efflux in
terol synthesis and cholesterol metabolism236–243. Studies renal cells294. The expression of LXR and target enzymes
in mice with HFD-induced obesity, ageing, or diabetes that regulate cholesterol efflux, including ATP-binding
have shown that increased SREBP‑2 expression and cassette sub-family A member 1 (ABCA1) and ABCG1,
activity are associated with increased cholesterol synthe- are decreased in human kidney biopsy samples from
sis and accumulation as well as the development of renal diabetic patients with ORG272,295 and in animal models
disease247–249,251,271. In obesity, as well as in diabetes and of diabetes251,296. Although the aetiology of the decrease
ageing, increased SREBP‑2 expression in the presence in LXR expression in the presence of high cellular cho-
of increased intracellular cholesterol indicates a break- lesterol is incompletely understood, lipopolysaccharide,
down of physiological feedback mechanisms. Potential TNF and IL‑1β downregulate LXR in kidney cells297.
Treatment of diabetic mice with LXR agonists reduces
cholesterol accumulation, decreases inflammatory medi-
↑ LDL receptor, ↑ oxidized LDL receptor, ↑ CD36 and ↑ SRA ators, and prevents progression of renal disease298–300. By
contrast, diabetic nephropathy is accelerated in LXR-
knockout mice; these mice exhibit increased cholesterol
↑ Cholesterol uptake
accumulation, inflammation, and oxidative stress300.
LXR activation also has additional effects in the kidney,
↑ Cholesterol ↑ Cholesterol ↓ Cholesterol including the reduction of renin activation301,302, and
synthesis efflux
inhibition of ENaC-mediated sodium transport 303 and
↓ ABCA1 and cystic fibrosis transmembrane conductance regulator
↑HMGCR ↓ Cholesterol catabolism ↓ ABCG1 (CFTR)-mediated chloride transport in collecting duct
cells304, which might have important roles in obesity-
↑ SREBP-2 ↓ LXR related kidney disease. In addition to LXR agonists,
↓ Bile acids induction of cholesterol efflux with cyclodextrin
↓ Bile acid
transporters LXR agonists improves renal disease in diabetic BTBR ob/ob mice295.

Figure 6 | Cholesterol metabolism in obesity-related glomerulopathy. Cholesterol Conclusions


Nature Reviews
accumulation can occur as a result of increased cholesterol synthesis mediated| Nephrology
by sterol ORG is defined pathologically as glomerulomegaly and
regulatory element-binding protein 2 (SREBP‑2) and its target enzymes including
FSGS occurring in patients with BMI ≥30 kg/m 2,
3‑hydroxy‑3‑methylglutaryl-coenzyme A reductase (HMGCR); increased cholesterol
uptake mediated by LDL receptor, oxidized LDL receptor, scavenger receptor A (SRA) or
and its prevalence is increasing in parallel with
CD36; decreased cholesterol efflux mediated by liver X receptor (LXR) and target enzymes; the worldwide obesity epidemic. Obesity induces
or decreased cholesterol catabolism mediated by bile acids and bile acid transporters. LXR increases in GFR, RPF, filtration fraction and tubular
agonists increase cholesterol efflux and decrease inflammation and endoplasmic reticulum reabsorption of sodium, promoting glomerular
stress. ABCA1, ATP-binding cassette subfamily A member 1; ABCG1, ATP-binding cassette hypertension. FSGS is mediated by the inability of
subfamily G member 1; HMGCR, hydroxymethylglutaryl-CoA reductase kinase. adaptive podocyte hypertrophy to keep pace with

14 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

glomerular tuft expansion, leading to podocyte failure. and small RCTs; however, larger prospective studies of
A predominance of perihilar FSGS and relatively mild longer duration are needed. Adipokines and ectopic lipid
foot process effacement helps to distinguish ORG from resulting from dysfunctional adipose tissue homeostasis
primary FSGS. Although subnephrotic proteinuria is in unhealthy obesity may lead to maladaptive changes
the most common presentation of ORG, less than half of of renal cells to cope with the mechanical forces of renal
patients have nephrotic-range proteinuria and the pres- hyperfiltration. This process may contribute to glomerular
ence of full nephrotic syndrome is distinctly unusual. The podocyte rarefaction, albuminuria, and eventually FSGS
clinical course is characterized by stable or slowly progres- and interstitial fibrosis. Lipid accumulation in the kid-
sive proteinuria, but up to one-third of patients develop ney (fatty kidney) has been associated with renal insulin
progressive renal failure and ESRD. Acquired reduction of resistance of podocytes, but not of tubular cells. Normal
renal mass and a low nephron endowment at birth have insulin/PI3K/Akt and mTOR signalling seems critical
been identified as predisposing factors for ORG. RAAS for normal podocyte function and adaptation. Altered
blockade is an effective therapeutic measure, but the fatty acid and cholesterol metabolism in obesity has an
antiproteinuric and renoprotective effects may be short- important role in lipid accumulation and regulation of
lived. Weight loss, either as a result of hypocaloric diets or inflammation, oxidative stress and fibrosis. SREBP antag-
bariatric surgery, has shown a consistent and remarkable onists and agonists of PPARα, FXR, TGR5 and LXR hold
antiproteinuric effect in case reports, retrospective studies promise for the treatment and prevention of ORG.

1. Coresh, J. et al. Prevalence of chronic kidney disease 16. Kambham, N., Markowitz, G. S., Valeri, A. M., Lin, J. & 27. Nagata, M. & Kriz, W. Glomerular damage after
in the United States. JAMA 298, 2038–2047 (2007). D’Agati, V. D. Obesity-related glomerulopathy: an uninephrectomy in young rats. II. Mechanical stress on
2. Farag, Y. M. & Gaballa, M. R. Diabesity: an overview emerging epidemic. Kidney Int. 59, 1498–1509 podocytes as a pathway to sclerosis. Kidney Int. 42,
of a rising epidemic. Nephrol. Dial. Transplant. 26, (2001). 148–160 (1992).
28–35 (2011). This study provided the first detailed clinical– 28. Tsuboi, N. et al. Low glomerular density with
3. Zammit, A. R., Katz, M. J., Derby, C., Bitzer, M. & pathologic analysis of ORG as contrasted with glomerulomegaly in obesity-related glomerulopathy.
Lipton, R. B. Chronic kidney disease in non-diabetic primary FSGS and demonstrates the rising Clin. J. Am. Soc. Nephrol. 7, 735–741 (2012).
older adults: associated roles of the metabolic prevalence of ORG diagnosed by kidney biopsy. 29. Hoy, W. E. et al. A stereological study of glomerular
syndrome, inflammation, and insulin resistance. PLoS 17. Praga, M. et al. Clinical features and long-term number and volume: preliminary findings in a
ONE 10, e0139369 (2015). outcome of obesity-associated focal segmental multiracial study of kidneys at autopsy. Kidney Int. 63,
4. Stenvinkel, P., Zoccali, C. & Ikizler, T. A. Obesity in glomerulosclerosis. Nephrol. Dial. Transplant. 16, S31–S37 (2003).
CKD — what should nephrologists know? J. Am. Soc. 1790–1798 (2001). 30. Hughson, M., Farris, A. B., 3rd, Douglas-Denton, R.,
Nephrol. 24, 1727–1736 (2013). This comprehensive clinical study describes the Hoy, W. E. & Bertram, J. F. Glomerular number and
5. Wickman, C. & Kramer, H. Obesity and kidney presenting clinical features and outcomes of ORG size in autopsy kidneys: the relationship to birth
disease: potential mechanisms. Semin. Nephrol. 33, in a large Spanish cohort. weight. Kidney Int. 63, 2113–2122 (2003).
14–22 (2013). 18. Hughson, M. D., Hoy, W. E., Douglas-Denton, R. N., 31. Luyckx, V. A. & Brenner, B. M. Birth weight, malnutrition
6. Guebre-Egziabher, F. et al. Ectopic lipid accumulation: Zimanyi, M. A. & Bertram, J. F. Towards a definition of and kidney-associated outcomes — a global concern.
a potential cause for metabolic disturbances and a glomerulomegaly: clinical-pathological and Nat. Rev. Nephrol. 11, 135–149 (2015).
contributor to the alteration of kidney function. methodological considerations. Nephrol. Dial. 32. Manalich, R., Reyes, L., Herrera, M., Melendi, C. &
Biochimie 95, 1971–1979 (2013). Transplant. 26, 2202–2208 (2011). Fundora, I. Relationship between weight at birth and
7. Nolan, E., O’Meara, Y. M. & Godson, C. Lipid 19. D’Agati, V. D., Kaskel, F. J. & Falk, R. J. Focal the number and size of renal glomeruli in humans: a
mediators of inflammation in obesity-related segmental glomerulosclerosis. N. Engl. J. Med. 365, histomorphometric study. Kidney Int. 58, 770–773
glomerulopathy. Nephrol. Dial. Transplant. 28 2398–2411 (2011). (2000).
(Suppl. 4), iv22–iv29 (2013). 20. D’Agati, V. D., Fogo, A. B., Bruijn, J. A. & 33. Bobulescu, I. A. et al. Triglycerides in the human
8. de Vries, A. P. et al. Fatty kidney: emerging role of Jennette, J. C. Pathologic classification of focal kidney cortex: relationship with body size. PLoS ONE
ectopic lipid in obesity-related renal disease. Lancet segmental glomerulosclerosis: a working proposal. 9, e101285 (2014).
Diabetes Endocrinol. 2, 417–426 (2014). Am. J. Kidney Dis. 43, 368–382 (2004). This human study is the first to show that obesity,
This review discusses the role of ectopic lipid 21. Chagnac, A. et al. Glomerular hemodynamics in severe as reflected by BMI, is associated with increased
accumulation in maladaptive changes of glomerular obesity. Am. J. Physiol. Renal Physiol. 278, triglyceride accumulation in the kidney,
cells in response to the haemodynamic forces of F817–F822 (2000). predominantly in tubular cells.
hyperfiltration and CKD. This human study shows that obesity-related 34. Wu, Y. et al. Obesity-related glomerulopathy: insights
9. GBD 2013 Risk Factors Collaborators. Global, glomerular hyperfiltration is mainly the result of an from gene expression profiles of the glomeruli derived
regional, and national comparative risk assessment of increase in transcapillary hydraulic pressure from renal biopsy samples. Endocrinology 147,
79 behavioural, environmental and occupational, and difference, mostly due to afferent arteriolar 44–50 (2006).
metabolic risks or clusters of risks in 188 countries, dilation. This study shows increased expression of multiple
1990–2013: a systematic analysis for the Global 22. Chen, H. M. et al. Podocyte lesions in patients with genes that regulate insulin function, lipid
Burden of Disease Study 2013. Lancet 386, obesity-related glomerulopathy. Am. J. Kidney Dis. metabolism, inflammation and fibrogenesis in
2287–2323 (2015). 48, 772–779 (2006). kidney biopsy samples from patients with ORG.
10. Ogden, C. L., Carroll, M. D., Kit, B. K. & Flegal, K. M. 23. Wharram, B. L. et al. Podocyte depletion causes Notably, it identified SREBP‑1, which mediates
Prevalence of childhood and adult obesity in the glomerulosclerosis: diphtheria toxin-induced podocyte fatty acid and triglyceride synthesis.
United States, 2011–2012. JAMA 311, 806–814 depletion in rats expressing human diphtheria toxin 35. D’Agati, V. D. Pathobiology of focal segmental
(2014). receptor transgene. J. Am. Soc. Nephrol. 16, glomerulosclerosis: new developments. Curr. Opin.
11. Wang, Y., Beydoun, M. A., Liang, L., Caballero, B. & 2941–2952 (2005). Nephrol. Hypertens. 21, 243–250 (2012).
Kumanyika, S. K. Will all Americans become 24. Wiggins, J. E. et al. Podocyte hypertrophy, 36. D’Agati, V. D. The spectrum of focal segmental
overweight or obese? Estimating the progression and ‘adaptation,’ and ‘decompensation’ associated with glomerulosclerosis: new insights. Curr. Opin. Nephrol.
cost of the US obesity epidemic. Obesity (Silver glomerular enlargement and glomerulosclerosis in the Hypertens. 17, 271–281 (2008).
Spring) 16, 2323–2330 (2008). aging rat: prevention by calorie restriction. J. Am. Soc. 37. Bhathena, D. B., Julian, B. A., McMorrow, R. G. &
12. Li, Q., Blume, S. W., Huang, J. C., Hammer, M. & Nephrol. 16, 2953–2966 (2005). Baehler, R. W. Focal sclerosis of hypertrophied
Ganz, M. L. Prevalence and healthcare costs of 25. Fukuda, A. et al. Growth-dependent podocyte failure glomeruli in solitary functioning kidneys of humans.
obesity-related comorbidities: evidence from an causes glomerulosclerosis. J. Am. Soc. Nephrol. 23, Am. J. Kidney Dis. 5, 226–232 (1985).
electronic medical records system in the United States. 1351–1363 (2012). 38. Hodgin, J. B., Rasoulpour, M., Markowitz, G. S. &
J. Med. Econ. 18, 1020–1028 (2015). This experimental study shows that the mTOR D’Agati, V. D. Very low birth weight is a risk factor for
13. Cohen, A. H. Massive obesity and the kidney. A pathway is important in the adaptive growth of the secondary focal segmental glomerulosclerosis. Clin.
morphologic and statistical study. Am. J. Pathol. 81, glomerulus in obesity and that FSGS can develop J. Am. Soc. Nephrol. 4, 71–76 (2009).
117–130 (1975). when the rate of podocyte hypertrophy is lower 39. Deegens, J. K. et al. Podocyte foot process effacement
14. Weisinger, J. R., Kempson, R. L., Eldridge, F. L. & than the rate of glomerular hypertrophy. as a diagnostic tool in focal segmental
Swenson, R. S. The nephrotic syndrome: a 26. Kriz, W., Hosser, H., Hahnel, B., Gretz, N. & glomerulosclerosis. Kidney Int. 74, 1568–1576 (2008).
complication of massive obesity. Ann. Intern. Med. 81, Provoost, A. P. From segmental glomerulosclerosis to 40. Ichikawa, I., Ma, J., Motojima, M. & Matsusaka, T.
440–447 (1974). total nephron degeneration and interstitial fibrosis: a Podocyte damage damages podocytes: autonomous
15. Chen, H. M. et al. Obesity-related glomerulopathy in histopathological study in rat models and human vicious cycle that drives local spread of glomerular
China: a case series of 90 patients. Am. J. Kidney Dis. glomerulopathies. Nephrol. Dial. Transplant. 13, sclerosis. Curr. Opin. Nephrol. Hypertens. 14,
52, 58–65 (2008). 2781–2798 (1998). 205–210 (2005).

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 15


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

41. D’Agati, V. Podocyte injury can be catching. J. Am. 61. Novick, A. C., Gephardt, G., Guz, B., Steinmuller, D. & 82. Navaneethan, S. D. et al. Weight loss interventions in
Soc. Nephrol. 22, 1181–1183 (2011). Tubbs, R. R. Long-term follow‑up after partial removal chronic kidney disease: a systematic review and meta-
42. Bonnet, F. et al. Excessive body weight as a new of a solitary kidney. N. Engl. J. Med. 325, 1058–1062 analysis. Clin. J. Am. Soc. Nephrol. 4, 1565–1574
independent risk factor for clinical and pathological (1991). (2009).
progression in primary IgA nephritis. Am. J. Kidney 62. Bosma, R. J., van der Heide, J. J., Oosterop, E. J., This systematic review and meta-analysis reportes
Dis. 37, 720–727 (2001). de Jong, P. E. & Navis, G. Body mass index is associated that in patients with CKD, surgical and non-surgical
This study was among the first to show that elevated with altered renal hemodynamics in non-obese healthy weight loss interventions significantly reduce
BMI is a risk factor for clinical and pathologic subjects. Kidney Int. 65, 259–265 (2004). proteinuria and blood pressure and seem to
progression of a glomerular disease other than ORG. 63. Praga, M. et al. Influence of obesity on the prevent further decline in renal function.
43. Hsu, C. Y., McCulloch, C. E., Iribarren, C., Darbinian, J. appearance of proteinuria and renal insufficiency after 83. Afshinnia, F., Wilt, T. J., Duval, S., Esmaeili, A. &
& Go, A. S. Body mass index and risk for end-stage unilateral nephrectomy. Kidney Int. 58, 2111–2118 Ibrahim, H. N. Weight loss and proteinuria: systematic
renal disease. Ann. Intern. Med. 144, 21–28 (2006). (2000). review of clinical trials and comparative cohorts.
This large historical cohort study of over 320,000 This study was the first to report detrimental Nephrol. Dial. Transplant. 25, 1173–1183 (2010).
adults demonstrated that higher BMI at baseline is effects of obesity in patients with renal mass 84. Bolignano, D. & Zoccali, C. Effects of weight loss on
an independent risk factor for subsequent ESRD in reduction. renal function in obese CKD patients: a systematic
multivariable analyses after adjustment for age, 64. Gonzalez, E. et al. Factors influencing the progression review. Nephrol. Dial. Transplant. 28 (Suppl. 4),
sex, race, education level, smoking status, cardiac of renal damage in patients with unilateral renal iv82–iv98 (2013).
disease, serum cholesterol, urinalysis proteinuria, agenesis and remnant kidney. Kidney Int. 68, 85. Praga, M. et al. Effects of body-weight loss and
hematuria, serum creatinine level, baseline blood 263–270 (2005). captopril treatment on proteinuria associated with
pressure level and presence of diabetes mellitus. 65. Whincup, P. H. et al. Birth weight and risk of type 2 obesity. Nephron 70, 35–41 (1995).
44. Sharma, S. G. et al. The modern spectrum of renal diabetes: a systematic review. JAMA 300, 86. Patil, M. R., Mishra, A., Jain, N., Gutch, M. &
biopsy findings in patients with diabetes. Clin. J. Am. 2886–2897 (2008). Tewari, R. Weight loss for reduction of proteinuria in
Soc. Nephrol. 8, 1718–1724 (2013). 66. White, S. L. et al. Is low birth weight an antecedent of diabetic nephropathy: comparison with angiotensin-
45. Serra, A. et al. Renal injury in the extremely obese CKD in later life? A systematic review of observational converting enzyme inhibitor therapy. Indian
patients with normal renal function. Kidney Int. 73, studies. Am. J. Kidney Dis. 54, 248–261 (2009). J. Nephrol. 23, 108–113 (2013).
947–955 (2008). 67. de Jong, F., Monuteaux, M. C., van Elburg, R. M., 87. Dixon, J. B. et al. Adjustable gastric banding and
Using protocol biopsies in patients undergoing Gillman, M. W. & Belfort, M. B. Systematic review and conventional therapy for type 2 diabetes: a randomized
bariatric surgery for morbid obesity, this study meta-analysis of preterm birth and later systolic blood controlled trial. JAMA 299, 316–323 (2008).
demostrated the presence of early pathologic pressure. Hypertension 59, 226–234 (2012). 88. Schauer, P. R. et al. Bariatric surgery versus intensive
abnormalities in patients lacking clinically 68. Mu, M. et al. Birth weight and subsequent blood medical therapy for diabetes — 3‑year outcomes.
detectable renal disease. pressure: a meta-analysis. Arch. Cardiovasc. Dis. 105, N. Engl. J. Med. 370, 2002–2013 (2014).
46. Goumenos, D. S. et al. Early histological changes in 99–113 (2012). 89. Chagnac, A. et al. The effects of weight loss on renal
the kidney of people with morbid obesity. Nephrol. 69. Vikse, B. E., Irgens, L. M., Leivestad, T., Hallan, S. & function in patients with severe obesity. J. Am. Soc.
Dial. Transplant. 24, 3732–3738 (2009). Iversen, B. M. Low birth weight increases risk for end- Nephrol. 14, 1480–1486 (2003).
47. D’Agati, V. D. & Markowitz, G. S Supersized kidneys: stage renal disease. J. Am. Soc. Nephrol. 19, This study shows that weight loss ameliorates
lessons from the preclinical obese kidney. Kidney Int. 151–157 (2008). obesity-related glomerular hyperfiltration,
73, 909–910 (2008). This important cohort study shows that low birth demonstrating the cause–effect relationship
48. Pehlivan, E. et al. Identifying the determinants of weight and intrauterine growth restriction increase between obesity and hyperfiltration.
microalbuminuria in obese patients in primary care the risk of ESRD. 90. Agrawal, V. et al. The effect of weight loss after
units: the effects of blood pressure, random plasma 70. Abitbol, C. L. et al. Obesity and preterm birth: bariatric surgery on albuminuria. Clin. Nephrol. 70,
glucose and other risk factors. J. Endocrinol. Invest. additive risks in the progression of kidney disease in 194–202 (2008).
39, 73–82 (2016). children. Pediatr. Nephrol. 24, 1363–1370 (2009). 91. Navaneethan, S. D. et al. Urinary albumin excretion,
49. Hashimoto, Y. et al. Metabolically healthy obesity and 71. Barker, D. J., Osmond, C., Forsen, T. J., Kajantie, E. & HMW adiponectin, and insulin sensitivity in type 2
risk of incident CKD. Clin. J. Am. Soc. Nephrol. 10, Eriksson, J. G. Trajectories of growth among children diabetic patients undergoing bariatric surgery. Obes.
578–583 (2015). who have coronary events as adults. N. Engl. J. Med. Surg. 20, 308–315 (2010).
50. Lin, W. Y. et al. Central obesity and albuminuria: both 353, 1802–1809 (2005). 92. MacLaughlin, H. L., Hall, W. L., Patel, A. G. &
cross-sectional and longitudinal studies in Chinese. 72. Andersen, L. G. et al. Birth weight, childhood body Macdougall, I. C. Laparoscopic sleeve gastrectomy is a
PLoS ONE 7, e47960 (2012). mass index and risk of coronary heart disease in novel and effective treatment for obesity in patients
51. Tsuboi, N. et al. Clinical features and long-term renal adults: combined historical cohort studies. PLoS ONE with chronic kidney disease. Obes. Surg. 22, 119–123
outcomes of Japanese patients with obesity-related 5, e14126 (2010). (2012).
glomerulopathy. Clin. Exp. Nephrol. 17, 379–385 73. Caballero, B. A nutrition paradox — underweight and 93. Neff, K. J. et al. The effect of bariatric surgery on renal
(2013). obesity in developing countries. N. Engl. J. Med. 352, function and disease: a focus on outcomes and
52. Chen, H. M. et al. Evaluation of metabolic risk marker 1514–1516 (2005). inflammation. Nephrol. Dial. Transplant. 28 (Suppl. 4),
in obesity-related glomerulopathy. J. Ren. Nutr. 21, 74. Mallamaci, F. et al. ACE inhibition is renoprotective iv73–iv82 (2013).
309–315 (2011). among obese patients with proteinuria. J. Am. Soc. 94. Reid, T. J. et al. The effect of bariatric surgery on renal
53. Praga, M. et al. Nephrotic proteinuria without Nephrol. 22, 1122–1128 (2011). function. Surg. Obes. Relat. Dis. 10, 808–813 (2014).
hypoalbuminemia: clinical characteristics and This post hoc analysis of the REIN trial shows that 95. Serra, A. et al. Long-term normal renal function after
response to angiotensin-converting enzyme inhibition. obesity predicts a higher risk of renal events but drastic weight reduction in patients with obesity-related
Am. J. Kidney Dis. 17, 330–338 (1991). ramipril treatment can abolish this excess risk. glomerulopathy. Obes. Facts 8, 188–199 (2015).
54. Praga, M. et al. Absence of hypoalbuminemia despite 75. Garg, R. & Adler, G. K. Aldosterone and the 96. Saleh, F., Kim, S. J., Okrainec, A. & Jackson, T. D.
massive proteinuria in focal segmental mineralocorticoid receptor: risk factors for Bariatric surgery in patients with reduced kidney
glomerulosclerosis secondary to hyperfiltration. Am. cardiometabolic disorders. Curr. Hypertens. Rep. 17, function: an analysis of short-term outcomes. Surg.
J. Kidney Dis. 33, 52–58 (1999). 52 (2015). Obes. Relat. Dis. 11, 828–835 (2015).
This study showed that patients with focal segmental 76. Bomback, A. S., Muskala, P., Bald, E., Chwatko, G. & 97. MacLaughlin, H. L. et al. Weight loss, adipokines, and
glomerulosclerosis secondary to hyperfiltration Nowicki, M. Low-dose spironolactone, added to long- quality of life after sleeve gastrectomy in obese patients
(including ORG) do not develop hypoalbuminemia term ACE inhibitor therapy, reduces blood pressure with stages 3–4 CKD: a randomized controlled pilot
even in the presence of nephrotic-range proteinuria. and urinary albumin excretion in obese patients with study. Am. J. Kidney Dis. 64, 660–663 (2014).
55. Sethi, S., Zand, L., Nasr, S. H., Glassock, R. J. & hypertensive target organ damage. Clin. Nephrol. 72, 98. Huan, Y., Tomaszewski, J. E. & Cohen, D. L. Resolution
Fervenza, F. C. Focal and segmental 449–456 (2009). of nephrotic syndrome after successful bariatric
glomerulosclerosis: clinical and kidney biopsy 77. Friedman, A. N., Chambers, M., Kamendulis, L. M. & surgery in patient with biopsy-proven FSGS. Clin.
correlations. Clin. Kidney J. 7, 531–537 (2014). Temmerman, J. Short-term changes after a weight Nephrol. 71, 69–73 (2009).
56. Sethi, S., Glassock, R. J. & Fervenza, F. C. Focal reduction intervention in advanced diabetic 99. Fowler, S. M. et al. Obesity-related focal and
segmental glomerulosclerosis: towards a better nephropathy. Clin. J. Am. Soc. Nephrol. 8, segmental glomerulosclerosis: normalization of
understanding for the practicing nephrologist. 1892–1898 (2013). proteinuria in an adolescent after bariatric surgery.
Nephrol. Dial. Transplant. 30, 375–384 (2015). 78. Saiki, A. et al. Effect of weight loss using formula diet Pediatr. Nephrol. 24, 851–855 (2009).
57. Shen, W. W. et al. Obesity-related glomerulopathy: on renal function in obese patients with diabetic 100. Lieske, J. C. et al. Kidney stones are common after
body mass index and proteinuria. Clin. J. Am. Soc. nephropathy. Int. J. Obes. (Lond.) 29, 1115–1120 bariatric surgery. Kidney Int. 87, 839–845 (2015).
Nephrol. 5, 1401–1409 (2010). (2005). 101. Turgeon, N. A. et al. The impact of renal function on
58. Nangaku, M. et al. In a type 2 diabetic nephropathy 79. Nicholson, A. S. et al. Toward improved management outcomes of bariatric surgery. J. Am. Soc. Nephrol.
rat model, the improvement of obesity by a low calorie of NIDDM: a randomized, controlled, pilot 23, 885–894 (2012).
diet reduces oxidative/carbonyl stress and prevents intervention using a lowfat, vegetarian diet. Prev. Med. 102. Brochner-Mortensen, J., Rickers, H. & Balslev, I. Renal
diabetic nephropathy. Nephrol. Dial. Transplant. 20, 29, 87–91 (1999). function and body composition before and after
2661–2669 (2005). 80. Morales, E., Valero, M. A., Leon, M., Hernandez, E. & intestinal bypass operation in obese patients. Scand.
59. Chander, P. N. et al. Nephropathy in Zucker diabetic Praga, M. Beneficial effects of weight loss in J. Clin. Lab. Invest. 40, 695–702 (1980).
fat rat is associated with oxidative and nitrosative overweight patients with chronic proteinuric 103. Ribstein, J., du Cailar, G. & Mimran, A. Combined
stress: prevention by chronic therapy with a nephropathies. Am. J. Kidney Dis. 41, 319–327 renal effects of overweight and hypertension.
peroxynitrite scavenger ebselen. J. Am. Soc. Nephrol. (2003). Hypertension 26, 610–615 (1995).
15, 2391–2403 (2004). 81. Straznicky, N. E. et al. Exercise augments weight loss 104. Scaglione, R. et al. Central obesity and hypertension:
60. Kawasaki, Y. et al. Two children with obesity-related induced improvement in renal function in obese pathophysiologic role of renal haemodynamics and
glomerulopathy identified in a school urinary metabolic syndrome individuals. J. Hypertens. 29, function. Int. J. Obes. Relat. Metab. Disord. 19,
screening program. Pediatr. Int. 56, 115–118 (2014). 553–564 (2011). 403–409 (1995).

16 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

105. Anastasio, P. et al. Glomerular filtration rate in 128. Rossi, G. P. et al. Body mass index predicts plasma 153. Song, J. et al. Regulation of blood pressure, the
severely overweight normotensive humans. Am. aldosterone concentrations in overweight-obese epithelial sodium channel (ENaC), and other key renal
J. Kidney Dis. 35, 1144–1148 (2000). primary hypertensive patients. J. Clin. Endocrinol. sodium transporters by chronic insulin infusion in rats.
106. Pecly, I. M. D., Genelhu, V. & Francischetti, E. A. Renal Metab. 93, 2566–2571 (2008). Am. J. Physiol. Renal Physiol. 290, F1055–F1064
functional reserve in obesity hypertension. Int. J. Clin. 129. Schorr, U., Blaschke, K., Turan, S., Distler, A. & (2006).
Pract. 60, 1198–1203 (2006). Sharma, A. M. Relationship between angiotensinogen, 154. Gesek, F. A. & Schoolwerth, A. C. Hormonal
107. Chagnac, A. et al. Obesity-induced glomerular leptin and blood pressure levels in young interactions with the proximal Na+-H+ exchanger. Am.
hyperfiltration: its involvement in the pathogenesis of normotensive men. J. Hypertens. 16, 1475–1480 J. Physiol. 258, F514–F521 (1990).
tubular sodium reabsorption. Nephrol. Dial. (1998). 155. Stenvinkel, P., Bolinder, J. & Alvestrand, A. Effects of
Transplant. 23, 3946–3952 (2008). 130. Achard, V., Boullu-Ciocca, S., Desbriere, R., Nguyen, G. insulin on renal haemodynamics and the proximal and
108. Wuerzner, G. et al. Marked association between & Grino, M. Renin receptor expression in human distal tubular sodium handling in healthy subjects.
obesity and glomerular hyperfiltration: a cross- adipose tissue. Am. J. Physiol. Regul. Integr. Comp. Diabetologia 35, 1042–1048 (1992).
sectional study in an African population. Am. J. Kidney Physiol. 292, R274–R282 (2007). 156. Sarafidis, P. A. & Bakris, G. L. The antinatriuretic effect
Dis. 56, 303–312 (2010). 131. Ehrhart-Bornstein, M., Arakelyan, K., Krug, A. W., of insulin: an unappreciated mechanism for
109. Deibert, P. et al. Acute effect of a soy protein-rich Scherbaum, W. A. & Bornstein, S. R. Fat cells may be hypertension associated with insulin resistance? Am.
meal-replacement application on renal parameters in the obesity–hypertension link: human adipogenic J. Nephrol. 27, 44–54 (2007).
patients with the metabolic syndrome. Asia Pac. factors stimulate aldosterone secretion from 157. Ogna, A. et al. Association between obesity and
J. Clin. Nutr. 20, 527–534 (2011). adrenocortical cells. Endocr. Res. 30, 865–870 (2004). glomerular hyperfiltration: the confounding effect of
110. Ahmed, S. B., Fisher, N. D., Stevanovic, R. & 132. Goodfriend, T. L. & Calhoun, D. A. Resistant smoking and sodium and protein intakes. Eur. J. Nutr.
Hollenberg, N. K. Body mass index and angiotensin- hypertension, obesity, sleep apnea, and aldosterone: 55, 1089–1097 (2016).
dependent control of the renal circulation in healthy theory and therapy. Hypertension 43, 518–524 158. King, A. J. & Levey, A. S. Dietary protein and renal
humans. Hypertension 46, 1316–1320 (2005). (2004). function. J. Am. Soc. Nephrol. 3, 1723–1737 (1993).
111. Porter, L. E. & Hollenberg, N. K. Obesity, salt intake, 133. Toke, A. & Meyer, T. W. Hemodynamic effects of 159. Lew, S. W. & Bosch, J. P. Effect of diet on creatinine
and renal perfusion in healthy humans. Hypertension angiotensin II in the kidney. Contrib. Nephrol. 135, clearance and excretion in young and elderly healthy
32, 144–148 (1998). 34–46 (2001). subjects and in patients with renal disease. J. Am. Soc.
112. Reisin, E., Messerli, F. G., Ventura, H. O. & 134. Arima, S. et al. Nongenomic vascular action of Nephrol. 2, 856–865 (1991).
Frohlich, E. D. Renal haemodynamic studies in obesity aldosterone in the glomerular microcirculation. J. Am. 160. Friedman, A. N. et al. Independent influence of dietary
hypertension. J. Hypertens. 5, 397–400 (1987). Soc. Nephrol. 14, 2255–2263 (2003). protein on markers of kidney function and disease in
113. Must, A. et al. The disease burden associated with 135. Ribstein, J., Du Cailar, G., Fesler, P. & Mimran, A. obesity. Kidney Int. 78, 693–697 (2010).
overweight and obesity. JAMA 282, 1523–1529 Relative glomerular hyperfiltration in primary 161. Chen, J. et al. The metabolic syndrome and chronic
(1999). aldosteronism. J. Am. Soc. Nephrol. 16, 1320–1325 kidney disease in U.S. adults. Ann. Intern. Med. 140,
114. Strazzullo, P. et al. Altered renal sodium handling in (2005). 167–174 (2004).
men with abdominal adiposity: a link to hypertension. 136. Kennedy, C. R. & Burns, K. D. Angiotensin II as a 162. Valensi, P. et al. Microalbuminuria in obese patients
J. Hypertens. 19, 2157–2164 (2001). mediator of renal tubular transport. Contrib. Nephrol. with or without hypertension. Int. J. Obes. Relat.
This investigation shows that increased abdominal 135, 47–62 (2001). Metab. Disord. 20, 574–579 (1996).
adiposity is associated with an enhanced rate of 137. Kawarazaki, W. et al. Angiotensin II- and salt-induced 163. Wennmann, D. O., Hsu, H.‑H. & Pavenstadt, H. The
proximal tubular sodium reabsorption, which might kidney injury through Rac1‑mediated renin–angiotensin–aldosterone system in podocytes.
have a role in the pathogenesis of glomerular mineralocorticoid receptor activation. J. Am. Soc. Semin. Nephrol. 32, 377–384 (2012).
hyperfiltration. Nephrol. 23, 997–1007 (2012). 164. Yano, Y. et al. Differential impacts of adiponectin on
115. Barbato, A. et al. Metabolic syndrome and renal 138. Granger, J. P. et al. Role of nitric oxide in modulating low-grade albuminuria between obese and nonobese
sodium handling in three ethnic groups living in renal function and arterial pressure during chronic persons without diabetes. J. Clin. Hypertens.
England. Diabetologia 47, 40–46 (2004). aldosterone excess. Am. J. Physiol. 276, R197–R202 (Greenwich) 9, 775–782 (2007).
116. Bickel, C. A., Verbalis, J. G., Knepper, M. A. & (1999). 165. Sharma, K. et al. Adiponectin regulates albuminuria
Ecelbarger, C. A. Increased renal Na‑K‑ATPase, NCC, 139. Hall, J. E., Granger, J. P., Smith, M. J. Jr & and podocyte function in mice. J. Clin. Invest. 118,
and β-ENaC abundance in obese Zucker rats. Am. Premen, A. J. Role of renal hemodynamics and arterial 1645–1656 (2008).
J. Physiol. Renal Physiol. 281, F639–F648 (2001). pressure in aldosterone ‘escape’. Hypertension 6, 166. Brenner, B. M. & Troy, J. L. Postglomerular vascular
117. Shah, S. & Hussain, T. Enhanced angiotensin I183–I192 (1984). protein concentration: evidence for a causal role in
II‑induced activation of Na+, K+-ATPase in the 140. Vaz, M. et al. Regional sympathetic nervous activity governing fluid reabsorption and glomerulotublar
proximal tubules of obese Zucker rats. Clin. Exp. and oxygen consumption in obese normotensive balance by the renal proximal tubule. J. Clin. Invest.
Hypertens. 28, 29–40 (2006). human subjects. Circulation 96, 3423–3429 (1997). 50, 336–349 (1971).
118. Brenner, B. M., Lawler, E. V. & Mackenzie, H. S. The 141. Davy, K. P. & Orr, J. S. Sympathetic nervous system 167. Ott, C. E., Haas, J. A., Cuche, J. L. & Knox, F. G. Effect
hyperfiltration theory: a paradigm shift in nephrology. behavior in human obesity. Neurosci. Biobehav. Rev. of increased peritubule protein concentration on
Kidney Int. 49, 1774–1777 (1996). 33, 116–124 (2009). proximal tubule reabsorption in the presence and
119. Bank, N. & Aynedjian, H. S. Progressive increases in 142. Esler, M. et al. Mechanisms of sympathetic activation absence of extracellular volume expansion. J. Clin.
luminal glucose stimulate proximal sodium absorption in obesity-related hypertension. Hypertension 48, Invest. 55, 612–620 (1975).
in normal and diabetic rats. J. Clin. Invest. 86, 787–796 (2006). 168. Ichikawa, I., Hoyer, J. R., Seiler, M. W. &
309–316 (1990). 143. Hall, J. E. et al. Obesity-induced hypertension: role of Brenner, B. M. Mechanism of glomerulotubular
120. Vallon, V., Blantz, R. C. & Thomson, S. Glomerular sympathetic nervous system, leptin, and melanocortins. balance in the setting of heterogeneous glomerular
hyperfiltration and the salt paradox in early J. Biol. Chem. 285, 17271–17276 (2010). injury. Preservation of a close functional linkage
[corrected] type 1 diabetes mellitus: a tubulo-centric 144. Young, C. N., Morgan, D. A., Butler, S. D., Mark, A. L. between individual nephrons and surrounding
view. J. Am. Soc. Nephrol. 14, 530–537 (2003). & Davisson, R. L. The brain subfornical organ microvasculature. J. Clin. Invest. 69, 185–198
121. Vallon, V., Richter, K., Blantz, R. C., Thomson, S. & mediates leptin-induced increases in renal sympathetic (1982).
Osswald, H. Glomerular hyperfiltration in activity but not its metabolic effects. Hypertension 61, 169. Weinbaum, S., Duan, Y., Satlin, L. M., Wang, T. &
experimental diabetes mellitus: potential role of 737–744 (2013). Weinstein, A. M. Mechanotransduction in the renal
tubular reabsorption. J. Am. Soc. Nephrol. 10, 145. Nasrallah, M. P. & Ziyadeh, F. N. Overview of the tubule. Am. J. Physiol. Renal Physiol. 299,
2569–2576 (1999). physiology and pathophysiology of leptin with special F1220–F1236 (2010).
Using an experimental diabetic model this study emphasis on its role in the kidney. Semin. Nephrol. 33, 170. Kriz, W. & Lemley, K. V. A potential role for mechanical
demonstrates that an increase in proximal tubular 54–65 (2013). forces in the detachment of podocytes and the
reabsorption of sodium leads to increased GFR by 146. Simonds, S. E. et al. Leptin mediates the increase in progression of CKD. J. Am. Soc. Nephrol. 26,
deactivation of tubuloglomerular feedback. blood pressure associated with obesity. Cell 159, 258–269 (2015).
122. Cappuccio, F. P., Strazzullo, P., Siani, A. & Trevisan, M. 1404–1416 (2014). This article reviews studies (most of which were
Increased proximal sodium reabsorption is associated 147. Greenfield, J. R. et al. Modulation of blood pressure performed in the authors’ laboratory) that
with increased cardiovascular risk in men. by central melanocortinergic pathways. N. Engl. demonstrate how mechanical forces associated
J. Hypertens. 14, 909–914 (1996). J. Med. 360, 44–52 (2009). with glomerular hypertension and hyperfiltration
123. Zingerman, B. et al. Effect of acetazolamide on obesity- 148. Tanida, M. et al. Effects of adiponectin on the renal result in glomerular enlargement and podocyte
induced glomerular hyperfiltration: a randomized sympathetic nerve activity and blood pressure in rats. detachment, leading to FSGS.
controlled trial. PLoS ONE 10, e0137163 (2015). Exp. Biol. Med. (Maywood) 232, 390–397 (2007). 171. Kriz, W., Elger, M., Mundel, P. & Lemley, K. V.
124. Engeli, S. et al. Weight loss and the renin– 149. Kinebuchi, S. et al. Short-term use of continuous Structure-stabilizing forces in the glomerular tuft.
angiotensin–aldosterone system. Hypertension 45, positive airway pressure ameliorates glomerular J. Am. Soc. Nephrol. 5, 1731–1739 (1995).
356–362 (2005). hyperfiltration in patients with obstructive sleep apnoea 172. Pereira, S. V. et al. Increased urine podocyte-
125. Tuck, M. L., Sowers, J., Dornfeld, L., Kledzik, G. & syndrome. Clin. Sci. (Lond.) 107, 317–322 (2004). associated messenger RNAs in severe obesity are
Maxwell, M. The effect of weight reduction on blood 150. Juncos, L. A. & Ito, S. Disparate effects of insulin on evidence of podocyte injury. Obesity (Silver Spring)
pressure, plasma renin activity, and plasma isolated rabbit afferent and efferent arterioles. J. Clin. 23, 1643–1649 (2015).
aldosterone levels in obese patients. N. Engl. J. Med. Invest. 92, 1981–1985 (1993). 173. Henegar, J. R., Bigler, S. A., Henegar, L. K., Tyagi, S. C.
304, 930–933 (1981). 151. Tucker, B. J. & Blantz, R. C. Effects of glomerular & Hall, J. E. Functional and structural changes in the
126. Cooper, R. et al. ACE, angiotensinogen and obesity: a filtration dynamics on the glomerular permeability kidney in the early stages of obesity. J. Am. Soc.
potential pathway leading to hypertension. J. Hum. coefficient. Am. J. Physiol. 240, F245–F254 (1981). Nephrol. 12, 1211–1217 (2001).
Hypertens. 11, 107–111 (1997). 152. Hayashi, K. et al. Effects of insulin on rat renal 174. Tobar, A. et al. Proximal tubular hypertrophy and
127. Bochud, M. et al. Plasma aldosterone is independently microvessels: studies in the isolated perfused enlarged glomerular and proximal tubular urinary
associated with the metabolic syndrome. Hypertension hydronephrotic kidney. Kidney Int. 51, 1507–1513 space in obese subjects with proteinuria. PLoS ONE 8,
48, 239–245 (2006). (1997). e75547 (2013).

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 17


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

This study demonstrates that glomerular 199. Musso, G. et al. Association of non-alcoholic fatty liver 222. Lennon, R. et al. Saturated fatty acids induce insulin
hyperfiltration in obese patients with proteinuria is disease with chronic kidney disease: a systematic review resistance in human podocytes: implications for
associated with proximal tubular epithelial and meta-analysis. PLoS Med. 11, e1001680 (2014). diabetic nephropathy. Nephrol. Dial. Transplant. 24,
hypertrophy and increased glomerular and tubular This meta-analysis reviews the impact of ectopic 3288–3296 (2009).
urinary space volume. These findings suggest that (liver) fat on CKD. 223. Sieber, J. et al. Regulation of podocyte survival and
the expanded glomerular and tubular urinary space 200. Hammer, S. et al. Metabolic imaging of human kidney endoplasmic reticulum stress by fatty acids. Am.
is a direct consequence of glomerular hyperfiltration. triglyceride content: reproducibility of proton J. Physiol. Renal Physiol. 299, F821–F829 (2010).
175. Ouchi, N., Parker, J. L., Lugus, J. J. & Walsh, K. magnetic resonance spectroscopy. PLoS ONE 8, 224. Welsh, G. I. et al. Insulin signaling to the glomerular
Adipokines in inflammation and metabolic disease. e62209 (2013). podocyte is critical for normal kidney function. Cell
Nat. Rev. Immunol. 11, 85–97 (2011). This human feasibility study is the first to Metab. 12, 329–340 (2010).
176. Cao, Y. Adipose tissue angiogenesis as a therapeutic non-invasively image kidney fat as a biomarker of This study shows the critical importance of insulin
target for obesity and metabolic diseases. Nat. Rev. fatty kidney. signalling for normal podocyte function.
Drug Discov. 9, 107–115 (2010). 201. Jonker, J. T.d. H. et al. Imaging fatty kidney using 225. Canaud, G. et al. AKT2 is essential to maintain
177. Kahn, C. R. Can we nip obesity in its vascular bud? proton MR spectroscopy: validation by porcine kidney podocyte viability and function during chronic kidney
Science 322, 542–543 (2008). biopsies. Nephrol. Dial. Transplant. 30, iii393–iii419 disease. Nat. Med. 19, 1288–1296 (2013).
178. Virtue, S. & Vidal-Puig, A. Adipose tissue (2015). This study shows the importance of Akt2, a
expandability, lipotoxicity and the metabolic syndrome 202. Berfield, A. K., Andress, D. L. & Abrass, C. K. downstream pathway of insulin signalling, in CKD
— an allostatic perspective. Biochim. Biophys. Acta IGF‑1‑induced lipid accumulation impairs mesangial with ORG-like features.
1801, 338–349 (2010). cell migration and contractile function. Kidney Int. 62, 226. Hale, L. J. et al. Insulin directly stimulates VEGF‑A
179. Briffa, J. F., McAinch, A. J., Poronnik, P. & 1229–1237 (2002). production in the glomerular podocyte. Am. J. Physiol.
Hryciw, D. H. Adipokines as a link between obesity and 203. Ruan, X. Z., Varghese, Z., Powis, S. H. & Renal Physiol. 305, F182–F188 (2013).
chronic kidney disease. Am. J. Physiol. Renal Physiol. Moorhead, J. F. Dysregulation of LDL receptor under 227. Laplante, M. & Sabatini, D. M. mTOR signaling in
305, F1629–F1636 (2013). the influence of inflammatory cytokines: a new growth control and disease. Cell 149, 274–293
180. Maisonpierre, P. C. et al. Angiopoietin‑2, a natural pathway for foam cell formation. Kidney Int. 60, (2012).
antagonist for Tie2 that disrupts in vivo angiogenesis. 1716–1725 (2001). 228. Godel, M. et al. Role of mTOR in podocyte function
Science 277, 55–60 (1997). 204. van Zonneveld, A. J. & Rabelink, T. J. Mesangial cells and diabetic nephropathy in humans and mice. J. Clin.
181. Schlondorff, D. & Banas, B. The mesangial cell defy LDL receptor paradigm. Kidney Int. 60, Invest. 121, 2197–2209 (2011).
revisited: no cell is an island. J. Am. Soc. Nephrol. 20, 2037–2038 (2001). This study demonstrates the important role of
1179–1187 (2009). 205. Li, Z. et al. Increased glomerular filtration rate in early mTOR in diabetic nephropathy.
182. Lindahl, P., Johansson, B. R., Leveen, P. & metabolic syndrome is associated with renal adiposity 229. Inoki, K. et al. mTORC1 activation in podocytes is a
Betsholtz, C. Pericyte loss and microaneurysm and microvascular proliferation. Am. J. Physiol. Renal critical step in the development of diabetic
formation in PDGF‑B‑deficient mice. Science 277, Physiol. 301, F1078–F1087 (2011). nephropathy in mice. J. Clin. Invest. 121, 2181–2196
242–245 (1997). This experimental study is the first to link renal (2011).
183. Singh, A. K. et al. Vascular factors altered in glucose- triglycerides to functional and microvascular 230. Tiwari, S. et al. Deletion of the insulin receptor in the
treated mesangial cells and diabetic glomeruli. hyperfiltration. proximal tubule promotes hyperglycemia. J. Am. Soc.
Changes in vascular factors impair endothelial cell 206. Chung, J. J. et al. Albumin-associated free fatty acids Nephrol. 24, 1209–1214 (2013).
growth and matrix. Lab. Invest. 84, 597–606 (2004). induce macropinocytosis in podocytes. J. Clin. Invest. 231. Meyer, C., Dostou, J., Nadkarni, V. & Gerich, J. Effects
184. Khairoun, M. et al. Early systemic microvascular 125, 2307–2316 (2015). of physiological hyperinsulinemia on systemic, renal,
damage in pigs with atherogenic diabetes mellitus 207. Clement, L. C. et al. Podocyte-secreted angiopoietin- and hepatic substrate metabolism. Am. J. Physiol.
coincides with renal angiopoietin dysbalance. PLoS like‑4 mediates proteinuria in glucocorticoid-sensitive 275, F915–F921 (1998).
ONE 10, e0121555 (2015). nephrotic syndrome. Nat. Med. 17, 117–122 (2011). 232. Mandel, L. J. Metabolic substrates, cellular energy
185. Kumar Vr, S. et al. Cathepsin S cleavage of protease- 208. Ruan, X. Z., Varghese, Z. & Moorhead, J. F. An update production, and the regulation of proximal tubular
activated receptor‑2 on endothelial cells promotes on the lipid nephrotoxicity hypothesis. Nat. Rev. transport. Annu. Rev. Physiol. 47, 85–101 (1985).
microvascular diabetes complications. J. Am. Soc. Nephrol. 5, 713–721 (2009). 233. Gerich, J. E. Role of the kidney in normal glucose
Nephrol. http://dx.doi.org/10.1681/ This is an important review of the updated homeostasis and in the hyperglycaemia of diabetes
ASN.2015020208 (2015). Moorhead hypothesis in nephrology. mellitus: therapeutic implications. Diabet. Med. 27,
186. Nielsen, R. et al. Endocytosis provides a major 209. Sun, Y. B. et al. Smad3 deficiency protects mice from 136–142 (2010).
alternative pathway for lysosomal biogenesis in kidney obesity-induced podocyte injury that precedes insulin 234. Marsenic, O. Glucose control by the kidney: an
proximal tubular cells. Proc. Natl Acad. Sci. USA 104, resistance. Kidney Int. 88, 286–298 (2015). emerging target in diabetes. Am. J. Kidney Dis. 53,
5407–5412 (2007). 210. Lin, H. M. et al. Transforming growth factor-β/Smad3 875–883 (2009).
187. Yaddanapudi, S. et al. CD2AP in mouse and human signaling regulates insulin gene transcription and 235. Mima, A. et al. Glomerular-specific protein kinase C‑β-
podocytes controls a proteolytic program that pancreatic islet β-cell function. J. Biol. Chem. 284, induced insulin receptor substrate‑1 dysfunction and
regulates cytoskeletal structure and cellular survival. 12246–12257 (2009). insulin resistance in rat models of diabetes and
J. Clin. Invest. 121, 3965–3980 (2011). 211. Yadav, H. et al. Protection from obesity and diabetes obesity. Kidney Int. 79, 883–896 (2011).
188. Faul, C. et al. The actin cytoskeleton of kidney by blockade of TGF-β/Smad3 signaling. Cell Metab. 236. Brown, M. S. & Goldstein, J. L. A proteolytic pathway
podocytes is a direct target of the antiproteinuric 14, 67–79 (2011). that controls the cholesterol content of membranes,
effect of cyclosporine A. Nat. Med. 14, 931–938 212. Kang, H. M. et al. Defective fatty acid oxidation in cells, and blood. Proc. Natl Acad. Sci. USA 96,
(2008). renal tubular epithelial cells has a key role in kidney 11041–11048 (1999).
189. Sever, S. et al. Proteolytic processing of dynamin by fibrosis development. Nat. Med. 21, 37–46 (2015). 237. Horton, J. D. Sterol regulatory element-binding
cytoplasmic cathepsin L is a mechanism for proteinuric This important study concludes that tubular fat is a proteins: transcriptional activators of lipid synthesis.
kidney disease. J. Clin. Invest. 117, 2095–2104 (2007). consequence of diminished fatty acid oxidation and Biochem. Soc. Trans. 30, 1091–1095 (2002).
190. Pinto-Sietsma, S. J. et al. A central body fat has a role in the development of kidney fibrosis. 238. Anderson, R. G. Joe Goldstein and Mike Brown: from
distribution is related to renal function impairment, 213. Nieth, H. & Schollmeyer, P. Substrate-utilization of the cholesterol homeostasis to new paradigms in
even in lean subjects. Am. J. Kidney Dis. 41, 733–741 human kidney. Nature 209, 1244–1245 (1966). membrane biology. Trends Cell Biol. 13, 534–539
(2003). 214. Wirthensohn, G. & Guder, W. G. Renal lipid metabolism. (2003).
191. Elsayed, E. F. et al. Waist‑to‑hip ratio, body mass Miner. Electrolyte Metab. 9, 203–211 (1983). 239. Horton, J. D. et al. Combined analysis of
index, and subsequent kidney disease and death. Am. 215. Stadler, K., Goldberg, I. J. & Susztak, K. The evolving oligonucleotide microarray data from transgenic and
J. Kidney Dis. 52, 29–38 (2008). understanding of the contribution of lipid metabolism knockout mice identifies direct SREBP target genes.
192. Fox, C. S. et al. Abdominal visceral and subcutaneous to diabetic kidney disease. Curr. Diab. Rep. 15, 40 Proc. Natl Acad. Sci. USA 100, 12027–12032
adipose tissue compartments: association with (2015). (2003).
metabolic risk factors in the Framingham Heart Study. 216. Shulman, G. I. Ectopic fat in insulin resistance, 240. Radhakrishnan, A., Sun, L. P., Kwon, H. J.,
Circulation 116, 39–48 (2007). dyslipidemia, and cardiometabolic disease. N. Engl. Brown, M. S. & Goldstein, J. L. Direct binding of
193. Einstein, F. H. et al. Differential responses of visceral J. Med. 371, 1131–1141 (2014). cholesterol to the purified membrane region of SCAP:
and subcutaneous fat depots to nutrients. Diabetes 217. Reaven, G. M. Banting lecture 1988. Role of insulin mechanism for a sterol-sensing domain. Mol. Cell 15,
54, 672–678 (2005). resistance in human disease. Diabetes 37, 259–268 (2004).
194. Kurella, M., Lo, J. C. & Chertow, G. M. Metabolic 1595–1607 (1988). 241. Engelking, L. J. et al. Schoenheimer effect explained
syndrome and the risk for chronic kidney disease 218. Oterdoom, L. H. et al. Fasting insulin modifies the — feedback regulation of cholesterol synthesis in mice
among nondiabetic adults. J. Am. Soc. Nephrol. 16, relation between age and renal function. Nephrol. mediated by Insig proteins. J. Clin. Invest. 115,
2134–2140 (2005). Dial. Transplant. 22, 1587–1592 (2007). 2489–2498 (2005).
195. Young, J. A. et al. Association of visceral and 219. Tomaszewski, M. et al. Glomerular hyperfiltration: a 242. Brown, M. S., Ye, J. & Goldstein, J. L. HDL miR‑ed
subcutaneous adiposity with kidney function. Clin. new marker of metabolic risk. Kidney Int. 71, down by SREBP introns. Science 328, 1495–1496
J. Am. Soc. Nephrol. 3, 1786–1791 (2008). 816–821 (2007). (2010).
196. de Vries, A. P. & Rabelink, T. J. A possible role of 220. ter Maaten, J. C. et al. Insulin’s acute effects on 243. Moon, Y. A. et al. The Scap/SREBP pathway is
cystatin C in adipose tissue homeostasis may impact glomerular filtration rate correlate with insulin essential for developing diabetic fatty liver and
kidney function estimation in metabolic syndrome. sensitivity whereas insulin’s acute effects on proximal carbohydrate-induced hypertriglyceridemia in animals.
Nephrol. Dial. Transplant. 28, 1628–1630 (2013). tubular sodium reabsorption correlation with salt Cell Metab. 15, 240–246 (2012).
197. Despres, J. P. & Lemieux, I. Abdominal obesity and sensitivity in normal subjects. Nephrol. Dial. 244. Sun, L., Halaihel, N., Zhang, W., Rogers, T. & Levi, M.
metabolic syndrome. Nature 444, 881–887 (2006). Transplant. 14, 2357–2363 (1999). Role of sterol regulatory element-binding protein 1 in
198. Stefan, N. & Haring, H. U. The role of hepatokines in 221. Hale, L. J. & Coward, R. J. Insulin signalling to the regulation of renal lipid metabolism and
metabolism. Nat. Rev. Endocrinol. 9, 144–152 kidney in health and disease. Clin. Sci. (Lond.) 124, glomerulosclerosis in diabetes mellitus. J. Biol. Chem.
(2013). 351–370 (2013). 277, 18919–18927 (2002).

18 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

245. Ruan, X. Z. et al. Regulation of lipoprotein trafficking 268. Wang, X. X. et al. The farnesoid X receptor modulates 289. Zheng, Y. et al. Anti-inflammatory effects of Ang-(1‑7)
in the kidney: role of inflammatory mediators and renal lipid metabolism and diet-induced renal in ameliorating HFD-induced renal injury through
transcription factors. Biochem. Soc. Trans. 32, 88–91 inflammation, fibrosis, and proteinuria. Am. J. Physiol. LDLr–SREBP2–SCAP pathway. PLoS ONE 10,
(2004). Renal Physiol. 297, F1587–F1596 (2009). e0136187 (2015).
246. Wang, Z. et al. Regulation of renal lipid metabolism, 269. Wang, X. X. et al. Diabetic nephropathy is accelerated 290. Gotoh, K. et al. Effects of hydrophilic statins on renal
lipid accumulation, and glomerulosclerosis in FVBdb/db by farnesoid X receptor deficiency and inhibited by tubular lipid accumulation in diet-induced obese mice.
mice with type 2 diabetes. Diabetes 54, 2328–2335 farnesoid X receptor activation in a type 1 diabetes Obes. Res. Clin. Pract. 7, e342–e352 (2013).
(2005). model. Diabetes 59, 2916–2927 (2010). 291. Calkin, A. C. & Tontonoz, P. Transcriptional integration
247. Jiang, T. et al. Diet-induced obesity in C57BL/6J mice 270. Hu, Z., Ren, L., Wang, C., Liu, B. & Song, G. Effect of of metabolism by the nuclear sterol-activated
causes increased renal lipid accumulation and chenodeoxycholic acid on fibrosis, inflammation and receptors LXR and FXR. Nat. Rev. Mol. Cell Biol. 13,
glomerulosclerosis via a sterol regulatory element- oxidative stress in kidney in high-fructose-fed Wistar 213–224 (2012).
binding protein‑1c‑dependent pathway. J. Biol. Chem. rats. Kidney Blood Press. Res. 36, 85–97 (2012). 292. Hong, C. & Tontonoz, P. Liver X receptors in lipid
280, 32317–32325 (2005). 271. Wang, X. X. et al. Vitamin D receptor agonist metabolism: opportunities for drug discovery. Nat.
248. Jiang, T., Liebman, S. E., Lucia, M. S., Li, J. & Levi, M. doxercalciferol modulates dietary fat-induced renal Rev. Drug Discov. 13, 433–444 (2014).
Role of altered renal lipid metabolism and the sterol disease and renal lipid metabolism. Am. J. Physiol. 293. Kratzer, A. et al. Synthetic LXR agonist attenuates
regulatory element binding proteins in the Renal Physiol. 300, F801–F810 (2011). plaque formation in apoE−/− mice without inducing
pathogenesis of age-related renal disease. Kidney Int. 272. Herman-Edelstein, M., Scherzer, P., Tobar, A., Levi, M. liver steatosis and hypertriglyceridemia. J. Lipid Res.
68, 2608–2620 (2005). & Gafter, U. Altered renal lipid metabolism and renal 50, 312–326 (2009).
249. Jiang, T., Liebman, S. E., Lucia, M. S., Phillips, C. L. & lipid accumulation in human diabetic nephropathy. 294. Wu, J. et al. Liver X receptor-α mediates cholesterol
Levi, M. Calorie restriction modulates renal expression J. Lipid Res. 55, 561–572 (2014). efflux in glomerular mesangial cells. Am. J. Physiol.
of sterol regulatory element binding proteins, lipid This study shows regulation of nuclear receptors, Renal Physiol. 287, F886–F895 (2004).
accumulation, and age-related renal disease. J. Am. transcription factors, and lipid metabolism 295. Merscher-Gomez, S. et al. Cyclodextrin protects
Soc. Nephrol. 16, 2385–2394 (2005). pathways in human diabetic nephropathy. podocytes in diabetic kidney disease. Diabetes 62,
250. Saito, K. et al. Lipid accumulation and transforming 273. Tanaka, Y. et al. Fenofibrate, a PPARα agonist, has 3817–3827 (2013).
growth factor-β upregulation in the kidneys of rats renoprotective effects in mice by enhancing renal 296. Tang, C., Kanter, J. E., Bornfeldt, K. E., Leboeuf, R. C.
administered angiotensin II. Hypertension 46, lipolysis. Kidney Int. 79, 871–882 (2011). & Oram, J. F. Diabetes reduces the cholesterol
1180–1185 (2005). 274. Hong, Y. A. et al. Fenofibrate improves renal exporter ABCA1 in mouse macrophages and kidneys.
251. Proctor, G. et al. Regulation of renal fatty acid and lipotoxicity through activation of AMPK-PGC‑1α in J. Lipid Res. 51, 1719–1728 (2010).
cholesterol metabolism, inflammation, and fibrosis in db/db mice. PLoS ONE 9, e96147 (2014). 297. Wang, Y., Moser, A. H., Shigenaga, J. K., Grunfeld, C.
Akita and OVE26 mice with type 1 diabetes. Diabetes 275. Li, L., Emmett, N., Mann, D. & Zhao, X. Fenofibrate & Feingold, K. R. Downregulation of liver X receptor-α
55, 2502–2509 (2006). attenuates tubulointerstitial fibrosis and inflammation in mouse kidney and HK‑2 proximal tubular cells by
252. Kume, S. et al. Role of altered renal lipid metabolism through suppression of nuclear factor-κB and LPS and cytokines. J. Lipid Res. 46, 2377–2387
in the development of renal injury induced by a high- transforming growth factor‑β1/Smad3 in diabetic (2005).
fat diet. J. Am. Soc. Nephrol. 18, 2715–2723 (2007). nephropathy. Exp. Biol. Med. (Maywood) 235, 298. Tachibana, H. et al. Activation of liver X receptor
253. Buga, G. M. et al. D-4F reduces EO6 383–391 (2010). inhibits osteopontin and ameliorates diabetic
immunoreactivity, SREBP‑1c mRNA levels, and renal 276. Kouroumichakis, I. et al. Fibrates: therapeutic nephropathy. J. Am. Soc. Nephrol. 23, 1835–1846
inflammation in LDL receptor-null mice fed a Western potential for diabetic nephropathy? Eur. J. Intern. (2012).
diet. J. Lipid Res. 49, 192–205 (2008). Med. 23, 309–316 (2012). 299. Kiss, E. et al. Lipid droplet accumulation is associated
254. Jun, H. et al. In vivo and in vitro effects of SREBP‑1 on 277. Effect of fenofibrate on progression of coronary-artery with an increase in hyperglycemia-induced renal
diabetic renal tubular lipid accumulation and RNAi- disease in type 2 diabetes: the Diabetes damage: prevention by liver X receptors. Am.
mediated gene silencing study. Histochem. Cell Biol. Atherosclerosis Intervention Study, a randomised J. Pathol. 182, 727–741 (2013).
131, 327–345 (2009). study. Lancet 357, 905–910 (2001). 300. Patel, M. et al. Liver X receptors preserve renal
255. Deji, N. et al. Structural and functional changes in the 278. Ansquer, J. C. et al. Fenofibrate reduces progression glomerular integrity under normoglycaemia and in
kidneys of high-fat diet-induced obese mice. Am. to microalbuminuria over 3 years in a placebo- diabetes in mice. Diabetologia 57, 435–446
J. Physiol. Renal Physiol. 296, F118–F126 (2009). controlled study in type 2 diabetes: results from the (2014).
256. Chin, H. J. et al. Omacor®, n-3 polyunsaturated fatty Diabetes Atherosclerosis Intervention Study (DAIS). 301. Morello, F. et al. Liver X receptors α and β regulate
acid, attenuated albuminuria and renal dysfunction Am. J. Kidney Dis. 45, 485–493 (2005). renin expression in vivo. J. Clin. Invest. 115,
with decrease of SREBP‑1 expression and triglyceride 279. Keech, A. et al. Effects of long-term fenofibrate therapy 1913–1922 (2005).
amount in the kidney of type II diabetic animals. on cardiovascular events in 9795 people with type 2 302. Kuipers, I. et al. Activation of liver X receptor-α
Nephrol. Dial. Transplant. 25, 1450–1457 (2010). diabetes mellitus (the FIELD study): randomised reduces activation of the renal and cardiac renin–
257. Wang, T. N. et al. SREBP‑1 mediates angiotensin controlled trial. Lancet 366, 1849–1861 (2005). angiotensin–aldosterone system. Lab. Invest. 90,
II‑induced TGF‑β1 upregulation and glomerular 280. Davis, T. M. et al. Effects of fenofibrate on renal 630–636 (2010).
fibrosis. J. Am. Soc. Nephrol. 26, 1839–1854 (2015). function in patients with type 2 diabetes mellitus: the 303. Soodvilai, S., Jia, Z., Fongsupa, S., Chatsudthipong, V.
258. Chen, G. et al. SREBP‑1 is a novel mediator of TGFβ1 Fenofibrate Intervention and Event Lowering in & Yang, T. Liver X receptor agonists decrease ENaC-
signaling in mesangial cells. J. Mol. Cell. Biol. 6, Diabetes (FIELD) Study. Diabetologia 54, 280–290 mediated sodium transport in collecting duct cells.
516–530 (2014). (2011). Am. J. Physiol. Renal Physiol. 303, F1610–F1616
259. Uttarwar, L., Gao, B., Ingram, A. J. & Krepinsky, J. C. 281. Group, A. S. et al. Effects of combination lipid therapy (2012).
SREBP‑1 activation by glucose mediates TGF-β in type 2 diabetes mellitus. N. Engl. J. Med. 362, 304. Raksaseri, P., Chatsudthipong, V., Muanprasat, C. &
upregulation in mesangial cells. Am. J. Physiol. Renal 1563–1574 (2010). Soodvilai, S. Activation of liver X receptors reduces
Physiol. 302, F329–F341 (2012). 282. Kim, D. et al. Metformin decreases high-fat diet- CFTR-mediated Cl− transport in kidney collecting duct
260. Hao, J. et al. PI3K/Akt pathway mediates high induced renal injury by regulating the expression of cells. Am. J. Physiol. Renal Physiol. 305, F583–F591
glucose-induced lipogenesis and extracellular matrix adipokines and the renal AMP-activated protein (2013).
accumulation in HKC cells through regulation of kinase/acetyl-CoA carboxylase pathway in mice. Int. 305. Berthoux, F., Mariat, C. & Maillard, N. Overweight/
SREBP‑1 and TGF‑β1. Histochem. Cell Biol. 135, J. Mol. Med. 32, 1293–1302 (2013). obesity revisited as a predictive risk factor in primary
173–181 (2011). 283. Decleves, A. E. et al. Regulation of lipid accumulation IgA nephropathy. Nephrol. Dial. Transplant. 28
261. Hao, J. et al. IFN-γ induces lipogenesis in mouse by AMP-activated kinase [corrected] in high fat diet- (Suppl. 4), iv160–iv166 (2013).
mesangial cells via the JAK2/STAT1 pathway. Am. induced kidney injury. Kidney Int. 85, 611–623 (2014). 306. Delanaye, P., Mariat, C., Cavalier, E. &
J. Physiol. Cell Physiol. 304, C760–C767 (2013). 284. Wang, X. X. et al. G protein-coupled bile acid receptor Krzesinski, J.‑M. Errors induced by indexing
262. Zhang, Y. J. et al. HMGB1/SREBP‑1 mediated IFN- TGR5 activation inhibits kidney disease in obesity and glomerular filtration rate for body surface area:
gamma-induced lipid deposition in mouse mesangial diabetes. J. Am. Soc. Nephrol. 27, 1362–1378 reductio ad absurdum. Nephrol. Dial. Transplant. 24,
cells. Zhongguo Ying Yong Sheng Li Xue Za Zhi 29, (2016). 3593–3596 (2009).
6–10 (in Chinese) (2013). This study was the first to show regulation of TGR5
263. Hao, J. et al. Phospho-mTOR: a novel target in in human ORG and diabetic nephropathy. Acknowledgements
regulation of renal lipid metabolism abnormality of 285. Zhang, Y. et al. Dysregulation of low-density The authors’ work is supported by the Dutch Kidney
diabetes. Exp. Cell Res. 319, 2296–2306 (2013). lipoprotein receptor contributes to podocyte injuries Foundation (IP11.56, A.P.V.), the Instituto de Salud Carlos III
264. Wang, H. et al. Co‑regulation of SREBP‑1 and mTOR in diabetic nephropathy. Am. J. Physiol. Endocrinol. (PI13/00342, Fondos FEDER and Redinren E.P.) and the
ameliorates lipid accumulation in kidney of diabetic Metab. 308, E1140–E1148 (2015). Fundacion Cajacanarias, project DIAB05 (E.P.).
mice. Exp. Cell Res. 336, 76–84 (2015). 286. Lhotak, S. et al. ER stress contributes to renal
265. Liu, W. et al. Phospho-GSK‑3β is involved in the high- proximal tubule injury by increasing Author contributions
glucose-mediated lipid deposition in renal tubular cells SREBP‑2‑mediated lipid accumulation and apoptotic All authors researched the data, discussed the content, wrote
in diabetes. Int. J. Biochem. Cell Biol. 45, 2066–2075 cell death. Am. J. Physiol. Renal Physiol. 303, the article and reviewed and/or edited the manuscript before
(2013). F266–F278 (2012). submission.
266. Hao, J. et al. PTEN ameliorates high glucose-induced 287. Yuan, Y. et al. Advanced glycation end products (AGEs)
lipid deposits through regulating SREBP‑1/FASN/ACC increase human mesangial foam cell formation by Competing interests statement
pathway in renal proximal tubular cells. Exp. Cell Res. increasing Golgi SCAP glycosylation in vitro. Am. The authors declare no competing interests.
317, 1629–1639 (2011). J. Physiol. Renal Physiol. 301,F236–F243 (2011).
267. Jiang, T. et al. Farnesoid X receptor modulates renal 288. Huang, W., Tang, L., Cai, Y., Zheng, Y. & Zhang, L.
lipid metabolism, fibrosis, and diabetic nephropathy. Effect and mechanism of the Ang-(1‑7) on human SUPPLEMENTARY INFORMATION
Diabetes 56, 2485–2493 (2007). mesangial cells injury induced by low density See online article: S1 (box)
This study is the first to show regulation and lipoprotein. Biochem. Biophys. Res. Commun. 450, ALL LINKS ARE ACTIVE IN THE ONLINE PDF
beneficial effects of FXR in the diabetic kidney. 1051–1057 (2014).

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 19


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like