ArticlePDF Available

Caveolin-1 in renal disease

Authors:

Abstract and Figures

p>Caveolin-1 is the essential structural formation for lipid raft formation. It has been ascribed to several disease processes in humans due to its ubiquitous distribution. Patients with chronic kidney disease suffer great morbidity and mortality where manipulation of caveolin-1 could lead to new potential therapeutic targets in this patient group. This review highlights caveolin-1 structure, signalling and provides examples of studies of caveolin-1 single nucleotide polymorphism in chronic kidney disease.</p
Content may be subject to copyright.
vv
Scienti c Journal of Genetics and Gene
Therapy
CC By
007
Citation: Chand S (2018) Caveolin-1 in renal disease. Scientific J Genet Gene Ther 4(1): 007-0014. DOI: http://dx.doi.org/10.17352/sjggt.000016
Clinical Group
http://dx.doi.org/10.17352/sjggt.000016DOI
Abstract
Caveolin-1 is the essential structural formation for lipid raft formation. It has been ascribed to several
disease processes in humans due to its ubiquitous distribution. Patients with chronic kidney disease suffer
great morbidity and mortality where manipulation of caveolin-1 could lead to new potential therapeutic
targets in this patient group. This review highlights caveolin-1 structure, signalling and provides examples
of studies of caveolin-1 single nucleotide polymorphism in chronic kidney disease.
Review Article
Caveolin-1 in renal disease
Sourabh Chand*
Renal Department, Shrewsbury and Telford NHS Trust,
UK
Received: 29 June, 2018
Accepted: 26 July, 2018
Published: 28 July, 2018
*Corresponding author: Sourabh Chand, Renal Depart-
ment, Shrewsbury and Telford NHS Trust, UK,
E-mail:
https://www.peertechz.com
Introduction
Chronic kidney disease (CKD) is a major global public health
issue that affects estimates of 10-16% of the general population
in developed countries leading to premature morbidity and
mortality [1]. In 2009-2010, the economic burden of chronic
kidney disease (CKD) on the English National Health Service
(NHS) was an estimated £1.45 billion (equivalent to 1.3% of
all NHS spending that year), with over half of the costs related
to the 2% of the CKD population with end-stage renal disease
(ESRD) requiring renal replacement therapy (RRT) [2]. The
resultant effect of the CKD burden is an excess in length of
stay in hospitals, hospital associated infections as well as an
excess of 7000 cerebral vascular events and 12000 myocardial
infarctions compared to age/gender matched controls [2].
Thus, it remains paramount to identify individuals with CKD at
the earliest time-point in order to instigate therapy to prevent
the progression to ESRD.
Caveolae are sub-class of non-clathrin coated lipid
rafts that were morphologically rst identi ed in 1953 via
transmission electron microscopy and appear as little cave-like
invaginations of the plasma membrane of 50-100 nanometres
in size [3]. In 1955, Yamada rst described caveolae existing in
the renal mouse glomerular capillary endothelium [4]. Caveolae
are ubiquitously distributed though predominately found in
endothelial cells, epithelial cells, striated and smooth muscle
cells, broblasts and type 1 pneumocytes, with almost twenty
percent of the total plasma membrane of adipocytes being
occupied by caveolae [5,6]. There are three main isoforms of
the caveolin protein, with caveolin-1 (CAV1) being essential for
the formation of caveolae. In 1999, caveolin-2 was described as
20-kDa protein that co-localises and is dependent on CAV1 to
form hetero-dimers [7] and caveolae membrane localisation to
the basolateral surface of epithelial cells, whilst caveolae with
CAV1 alone, are not seen on the apical surface [8]. Caveolin-3
has a similar morphology to caveolin-1, and is mostly found in
cardiac, smooth and skeletal myocytes [9].
Caveolae are largely composed of cholesterol with
concentrated glycosphingolipids and sphingomyelin relative
to the plasma membrane distribution, with CAV1 required
for its structural stability. As shown in gure 1, caveolae can
form either the classical omega or little cave, enlongated (for
channel formation) or a direct vesicle formation mostly at the
basolateral surface [10]. This depends on the proteins called
cavins whose abundance changes the mobility of CAV1 and
thus structural integrity of the caveolar structure allowing the
structure and its contents to be endocytosed. If the ratio of
cavin-1 to cavin-2 is higher, then the caveolae omega shape
DŽĚŝĮĞd from RothbĞrg Ğt al (10) and ChidloǁĞƚĂů(11). Cav-1, cavĞŽůŝŶ-1; Cav-Ϯ͕ĐĂǀĞolin-2. Bar on
ĞůĞctron micrograph is 25 micromĞƚĞƌƐ
Figure 1: Electron micrograph and schematic representation of caveolae
structures.
008
Citation: Chand S (2018) Caveolin-1 in renal disease. Scientific J Genet Gene Ther 4(1): 007-014. DOI: http://dx.doi.org/10.17352/sjggt.000016
is favoured; if cavin-2 expressed levels higher than cavin-1,
then the elongated shape is formed whilst cavin-3 expression
directs vesicle formation [11]. Depletion of cholesterol results in
decreased CAV1 expression that can also lead to destabilisation
of the caveolae structure to become mobile from the plasma
membrane.
Caveolin-1 is a protein that remains intracellular, formed
via the endoplasmic reticulum (ER) system and transported
from the Golgi apparatus to the plasma membrane. CAV1
exit from the Golgi apparatus is accelerated by increasing
cholesterol and inhibited after glycosphingolipid depletion
[12]. There are two isoforms of CAV1, alpha of 178 amino acid
length and beta which is 31 amino acid lengths shorter, with
the former isoform having a higher af nity for the plasma
membrane. The N’ terminal and C’ termini face the cytoplasm,
after tyrosine phosphorylation and palmitoylation respectively.
Between the termini, there is a hairpin structure hydrophobic
domain of 32 amino acids (residues 102-134) that inserts to
plasma membrane and is involved in hetero-oligomerization of
CAV1 with caveolin-2 [13]. Residues 82-101 and 135-150 ank
this region, with the latter termed C’ membrane attachment
domain that contains a cis-Golgi targeting sequence. The N’
membrane attachment domain or caveolin scaffolding domain
(residue 82-101) is integral in membrane localisation and
anchoring of various proteins within caveolae and regulation
(both inhibition and enhancement) of their signalling activity
[5]. Another important site is the tyrosine-14 residue (Y14),
which requires phosphorylation for caveolar endocytosis and is
important in cell adhesion.
Caveolin-1 gene
The human caveolin-1 gene is located on the long arm
of chromosome 7, at genomic locus 7q31.1 (7:116540796) as
shown in gure 2. It consists of 3 exons (30, 165 and 342 base
pair in length respectively) and 2 introns (1.5 and 32 kilobases
in length) spaced in-between. The rst exons contain CpG
islands that via methylation are thought to be the main part
of CAV1 gene expression in cancer cell lines [14]. The third
exon harbours the functional oligomerisation, scaffolding,
transmembrane and C’ membrane attachment domains that
are highly conserved across species [15].
Caveolin-1 signalling
There are several roles of caveolae and CAV1 which are
integral to cellular function. The most common involves
vesicular transport of macromolecules (such as albumin) via
transcytosis from the luminal side of the capillary endothelium
to the interstitial space via membrane-bound vesicles. The
accumulation of gold-labelled albumin in the interstitial
space is not seen in the knockout CAV1 mouse by transmission
electron microscopy as compared to the wild-type mouse [16].
Endocytosis is the second example of vesicular membrane
traf cking by caveolae. Caveolae share similar vesicle docking
and fusion molecules (soluble N-ethylmaleimide-sensitive
factor attachment protein receptors (SNARE) and dynamin)
as seen in the traditional clathrin-mediated endocytosis.
Pathogens such as Simian virus 40 and the cholera toxin utilise
collections of caveolae for internalisation into the cell, forming
with a distinct endosomal compartment with a neutral pH
called a caveosome, for delivery to the ER and Golgi apparatus
with recycling of CAV1 to the plasma membrane [5,17].
CAV1 and caveolae are also integral to cellular signalling
and signal transduction by compartmentalising receptors upon
ligand binding, acting as a chaperone of signalling molecules
and ligand bound receptors for delivery to the cell nucleus as
well as concentrating such events in the con nes of a speci c
subcellular environment [5]. Receptors such as the epidermal
growth factor receptor (EGFR) and transforming growth factor
beta (TGF) receptor are internalised by caveolae upon ligand
binding after phosphorylation of Y14 part of CAV1 [18]. This
internalisation is dependent on Src kinases and protein kinase
C [19]. Golgi derived CAV1 via SNARE protein syntaxin-6 is
also essential for plasma membrane delivery of receptors
angiotensin II type 1, insulin and the stretch activated channel
short transient receptor potential channel-1 [12].
In the majority, CAV1 effects are inhibitory upon cellular
signalling, acting to degrade receptors except in the case of the
insulin receptors. The main outcomes of CAV1 signal association
are pro-apoptotic and anti- brotic. The most commonly
described signalling CAV1 associations are with endothelial
nitric oxide synthase (eNOS) and TGF pathways. CAV1 via
its CSD binds eNOS resulting in its inactivity and can act as a
reservoir of inactivated eNOS. The in ux of calcium leads to
more calmodulin recruitment and binding to the eNOS enzyme
leading to its release from bound CAV1 and thus restoration of
electron ow from NADPH to its reductase domain avins and
then to the heme moiety of eNOS, generating the vasodilator
nitric oxide (NO) from L-arginine. The generation of NO leads to
a dissociation of CAV1 scaffold, leading to termination of signal
transduction. However, if the substrate L-arginine becomes
limited, eNOS functions in an uncoupled mode, leading to the
electron transfer to the heme moiety to react with oxygen to
form reactive oxygen species (ROS) [20].
ROS releases TGF-1 from its latency-associated peptide
and latent TGF-binding protein to become activated [21]. Upon
this ligand binding, TGF type 1 receptor is phosphorylated
after binding with TGF type II receptor to form a serine/
threonine kinase heterotetrameric complex (Figure 3). If this
complex is internalised by the early-endosome antigen 1 non-
lipid/clathrin raft pathway, this phosphorylates the SARA/
Smad 2/3 complex downstream to induce a conformational
change that results in heteromerization of Smad 4 and
translocation to the nucleus to regulate transcription of pro-
brotic target genes as well as the downregulation of CAV1
The red rectangle shows the locaƟon of the caveolin-1 gene on chromosome 7
Figure 2: The caveolin-1 human gene on chromosome 7 and its three exons below
with size in base pairs (modi ed from (5)).
009
Citation: Chand S (2018) Caveolin-1 in renal disease. Scientific J Genet Gene Ther 4(1): 007-014. DOI: http://dx.doi.org/10.17352/sjggt.000016
and Smad 7 [22]. However, if the TGF receptor I/II complex is
internalised by caveolae, then SMURF/Smad 7 is recruited and
exerts a negative effect upon TGF signalling by proteasomal
degradation and ubiquitination of the TGF receptor complex
in CAV1 positive vesicles. In addition, CAV1 diminishes Smad
2 phosphorylation, disrupts its interaction with Smad 4, and
prevents Smad 2 translocation to the nucleus, further reducing
TGF signalling [22].
Other signalling events associated with caveolae endocytosis
involve bronectin degradation and integrin internalisation
[23,24]. The detachment of broblasts triggers ganglioside
GM1 internalisation via CAV1 and its Y14 phosphorylation; GM1
internalisation leads to Rac1 loss from the plasma membrane
and its reduced activation [12]. Thus not only is CAV1 important
in compartmentalising signal transduction in caveolae by
concentrating and localising signalling molecules by acting as
docking points for numerous cell surface receptors after ligand
binding, but also regulates cell adhesion, cell migration via 1-
integrins, extracellular matrix (ECM) interactions and acts as
ow sensors in endothelial cells and regulates stretch-induced
cell cycle progression [12,24]. CAV1 response to chronic shear
stress is to increase its levels with redistribution from the
Golgi complex to the plasma membrane and formation of
caveolae [25]. This leads to increased mechanosensitivity with
activation of signalling pathways such as eNOS and mitogen-
activated protein kinase (MAPK). With Cav1-/- mice, they
exhibit defects in chronic ow-dependent remodelling of blood
vessels [26]. Upon stretch, cell cycle progression is inhibited
with CAV1 downregulation or in Cav1-/- smooth muscle cells
through pathways including PI3 kinase-AKT/protein kinase B,
MAPK ERK, c-Src and integrins [27].
Caveolin-1 in renal adverse outcomes
Ca veolin-1 in renal brosis: As kidney function deteriorates
in CKD, more interstitial brosis is found amongst patients
irrespective of their underlying cause of CKD. CAV1 has been an
attractive protein to investigate in the context of organ brosis
as suggested with its effects described in the previous section
and the following section summarises renal brotic CAV1
research [28].
CAV1 interaction with TGF in brosis is a common
pathway by which renal and non-renal chronic disease
progresses and thus has been extensively investigated. Li et
al. used primary murine pulmonary endothelial cells that were
capable of obtaining a myo broblast phenotype after TGF-1
stimulation, inducing SMA expression from wild-type cells.
Cav1-/- cells produced very high spontaneous levels of SMA
that was corrected upon CAV1 functional restoration using its
CSD peptide; thus CAV1 regulates endothelial-to-mesenchymal
transition in tissue brosis [29]. Ito et al. [30], showed that
adding hyaluronan with its receptor CD44 to immortalised renal
proximal tubular epithelial cells (HK2) led to an increase of
lipid raft internalisation of TGF via MAP kinase from the non-
lipid pathway predominance. The authors co-cultured HK2
cells with cells transfected with a Smad responsive promoter
and blocked several components (e.g. CD44 and its interaction
with hyaluronan) along this pathway to con rm their nding.
As the authors state, this con icts with previous work with
metastatic breast tumour cells that suggest hyaluronan
promoted the non-lipid pathway, and thus the nding may be
dependent on speci c tissue microenvironments. Whilst Zhang
et al. [31], have shown stimulation with IL-6 led to more non-
lipid raft pathway dominance for the TGF receptor in HK2
cells and a decrease in the association of the IL-6 receptor
to CAV1 upon co-immunoprecipitation. This was after the
observation that IL-6 is only expressed on proximal tubular
epithelial cells at times of renal disease. However despite the
af nity labelling ndings, it was noted that stimulation with
IL-6 and TGF-1 did not lead to an increase in Smad 2 or 3 or
7 protein expression, but only the luciferase assay suggested
more Smad activity.
As well as TGF, there have been many other signalling
events associated with CAV1 in renal brosis. Peng et al [32],
showed a Src dependent phosphorylation of the CAV1 Y14
site occurred with RhoA activation and formation of ROS
when they mechanically stretched mesangial cells via a cyclic
vacuum as a model of intraglomerular hypertension leading to
glomerulosclerosis. In the Cav1 knockout mouse, there was no
RhoA or ROS activation but this function was promptly restored
upon restoration of Cav1 [33].
As another model of hypertension, Bocanegra et al. [34],
investigated the protective pro le of losartan upon proximal
tubular cells from spontaneously hypertensive rats, whose
CAV1 expression was signi cantly reduced compared to
control. Losartan (an angiotensin II type 1 receptor antagonist)
was noted to reduce ROS through Nox4 downregulation and
NADPH inactivation, through increased levels of CAV1 and co-
localisation with Heat shock protein 70. However two questions
remain: was this just a by-product of the blood pressure lowering
ability of losartan and why none of these effects were seen in
Figure 3: Schematic representation of TGFβ ligand (diamond yellow) receptor
complex I and II internalisation via clathrin and caveolae rafts (modi ed from (28))
010
Citation: Chand S (2018) Caveolin-1 in renal disease. Scientific J Genet Gene Ther 4(1): 007-014. DOI: http://dx.doi.org/10.17352/sjggt.000016
the control group? The editorial comment focussed on the
mechanisms behind hypertension and ROS production causing
reduced renal function and structural damage; this was mainly
to inappropriate activation of intrarenal angiotensin II within
the proximal tubules due to its translocation of angiotensin
II type 1 receptor (AT1) from the plasma membrane to CAV1
rich domains. Here, CAV1 acts as a molecular chaperone to
AT1, providing a platform for redox signalling events through
NADPH oxidase dependent production of ROS via Nox4 [35].
Genetically obese Zucker rats on a casein diet have also
been shown to have reduced eNOS and raised CAV1 in renal
structural damaged kidneys as compared to those on a soy diet,
but again, CAV1 is known to uctuate with cholesterol levels
making it dif cult to interpret the ndings [36]. From Valles et
al. [37], ureteric obstruction leads to an early rapid intrarenal
angiotensin II rise that leads to an increase in extracellular
matrix protein production and oxidative stress damage causing
brosis. Nitric oxide can ameliorate interstitial brosis and
in loss of eNOS leads to marked tubulointerstitial brosis
in the inducible NOS knockout mouse. CAV1 is expressed in
vascular endothelium, smooth muscle and epithelial cells
of the proximal, distal and convoluted tubules and ducts.
CAV1 also leads to apoptosis via inhibition of p42/44 MAP
kinase signalling. Thus the authors wished to look at the
role of CAV1 expression in congenital unilateral ureteropelvic
junction obstruction with grade IV hydronephrosis on imaging
requiring surgical intervention after vesico-ureteral re ux
was excluded. The 19 children had ‘normal renal function for
their age’, were on no medication and were normotensive. The
scorers of interstitial volume and CAV1 staining were blinded.
There were two groups with group 1 split into subset A with
obstruction <1yr, subset B with obstruction >1yr, and group 2
had signi cant renal impairment compared to group 1 (99mTc-
DPTA renal scan expression of kidney ltration rate 28.8±2%
to 39.7±2.1% respectively). The results showed group 2 CAV1
was present in the proximal tubule unlike control and group 1,
as well as co-localising with the AT1 receptor. Increased CAV1
expression was con rmed on Western Blotting for protein
transcription from group 2’s urine. eNOS was low in group2
(p<0.01). Unfortunately the control was based on children with
renal cell carcinoma where it is known that CAV1 has expression
can be altered and may thus not act as a true control.
Further to the above human study suggesting increased
CAV1 expression in the more chronic and brotic group in
children, histological increased glomerular expression of CAV1
in Japanese patients has been identi ed in diseases that target
the glomerulus such as diabetic nephropathy, membranous
nephropathy and focal segmental glomerulosclerosis [38].
It was also noted that CAV1 expression was also reduced
in glomerular endothelial cells with the use of steroids. In
diabetic nephropathy, murine Cav1 knockout led to a worse
glomerulosclerosis and albuminuria in the streptozotocin
model of type 1 diabetic nephropathy, though there is limited
tubulointerstitial brosis in this model [39]. CAV1 increased
expression was also noted in a streptozotocin rat model
of diabetic nephropathy with increased VEGF receptor 2
(VEGFR2)/CAV1 association in vivo [40]. In vitro, primary rat
mesangial cells after stimulation with VEGF, caused more CAV1/
VEGFR2, CAV1/Src expression and bronectin upregulation
via RhoA activation. Transfection of mesangial cells with an
overexpression of non-phosphorylatable CAV1 Y14 prevented
VEGF-induced RhoA activation and bronectin upregulation.
Another model of nodular glomerulosclerosis utilises human
urinary free light chains from patients suffering from light chain
deposition disease injecting to wild-type and Cav1 knockout
rats’ tail veins to induce nodular glomerulosclerosis. In the
Cav1 knockout, there was increased nodular glomerulosclerosis
and increased mesangial matrix production, however there was
a mix of gender used that may have in uenced the results [41].
Other human studies have focussed on CAV1 in renal
transplantation. Yamamoto et al. [42], have previously
shown that de novo caveolae formation occurs in transplant
glomerulopathy in glomerular endothelial cells and now
investigate CAV1 in chronic active antibody mediated rejection
and thus transplant capillaropathy, where they found CAV1
to be associated with peritubular capillary endothelia that is
not normally present in healthy kidney. Pontrelli et al. [43],
also investigated chronic allograft nephropathy secondary
to immune system activation where CD40 is upregulated in
acute rejection in proximal tubular epithelial cells. This led to
increased Lyn phosphorylation and thus NFҡB activation. Lyn
phosphorylation is strictly associated with CAV1 and inhibiting
Lyn blocked the pro brotic induction of PAI-1.
Park et al. [44], have used an unilateral ureteric obstruction
(UUO) model in FVB/N mice to investigate the surge of stem
cells that occurs upon 10 days after ligation of the left ureter. In
the Cav1 knockout, the mesenchymal stem cell post obstructive
surge was blunted signi cantly which led to no regeneration of
the parenchyma and marked brosis as measured by Sirius red
staining. Indeed some of the surge could be from the response
of the resident kidney stem cells. In this model, the FVB/N
wild-type mice are a different strain to the Cav1 knockout mice
and the control kidney was the contralateral kidney of each
mouse. These factors could affect the interpretation of their
ndings as different mouse strains have varying susceptibility
to brosis and the contralateral compensation after ligation of
the ureter could be different.
Chand et al. [45], also investigated the UUO model at day 3
and day 14 with sham operated mice being the same age, strain
and gender as mice undergoing UUO as their Cav1 knockout
mice. They showed that there was a more profound brosis
at day 14 of the UUO model with the Cav1 knockout mice than
their wild-type counterparts, but at day 3 the wild-type had
more brosis. The main difference was the abundance of F4/80
positive stained cells on confocal microscopy of frozen kidney
sections in the wild-type compared to the Cav1 knockout mice.
In summary, there appears to be negative effect of CAV1
expression leading to a worse renal phenotype especially in
glomerular disease. However, this is in opposition to the non-
renal literature where the CAV1 reduction in patient samples
or knockout of caveolin-1 in vitro has been found to lead to
a more brotic phenotype. This may be due to the organ and
microenvironment studied such as in bleomycin induced lung
011
Citation: Chand S (2018) Caveolin-1 in renal disease. Scientific J Genet Gene Ther 4(1): 007-014. DOI: http://dx.doi.org/10.17352/sjggt.000016
brosis [46], idiopathic pulmonary brosis [47], scleroderma/
systemic sclerosis [48], cardiac brosis [49], and keloid scars
[50], which involve more TGF dependent processes that are
reliant on the expression of CAV1 in broblasts.
Pleiotr opic effects of caveolin-1 in adverse renal out-
comes
As well as tubulointerstitial brosis, there are common
adverse outcomes experienced by patients with CKD as
their renal disease progresses. Due its ubiquitous nature of
distribution, CAV1 has been associated with many of these
outcomes.
Infection: In patients suffering from scleroderma associated
lung disease, their bronchoalveolar lavage revealed activated
monocytes and polymorphonuclear cells; these patients’
monocytes, neutrophils and T cells had a reduced expression
of CAV1 [50]. In mice exposed to lipopolysaccharide challenge,
the deletion of Cav1 led to a decreased expression of CD14
and CD36 during macrophage differentiation and suppressed
phagocytic ability and impaired bacterial clearance [51]. Cav1
knockout mice also had an increased mortality with induced
Pseudomonas aeruginosa and Klebsiella pneumoniae sepsis
[52,53]. In cystic brosis, effective internalisation of pathogens
such as Pseudomonas aeruginosa is required for an appropriate
immune response for its clearance; its internalisation is reliant
on CAV1 in type 1 pneumocytes and bladder epithelium [54].
CAV1 interaction with protein kinase C upon activation and
calcium release is essential for its translocation to caveolae
in order for production of infectious enveloped human
cytomegalovirus particles in broblasts [55]. Infection with
polyomavirus viraemia can cause BK nephropathy in renal
transplant recipients. Moriyama et al. in vitro data shows this
viral entry in human proximal tubular epithelial cells requires
co-localisation with CAV1 for caveolae entry into the cells [56].
Cardiovascular disease: CAV1 has been associated with
an in ammatory macrophage phenotype that promotes
atherosclerosis by the production of foam cells [57]. Schwencke
et al. have found reduced CAV1 expression in VSMC of human
atheroma [58]. CAV1 also binds eNOS in an inactive state and
with an in ux of calcium, its release thus affecting vascular
function [59]. In the Cav1 knockout mouse, cardiac hypertrophy
occurs despite the high presence of caveolin-3, in which the
latter is thought to be the predominant isoform of caveolin in
the heart [60].
Malignancy: CAV1 may have a tumour suppressive role
depending on the cancer type. CAV1 has been shown to promote
apoptosis and in Cav1 knockout mice, hyperactivation of the
p42/44 MAP kinase cascade and cyclin D1 upregulation in breast
cancer models lead to cancer progression and metastases [61].
CAV1 has been considered as a prognostic marker in several
cancers [62]. However, in prostate cancer, CAV1 upregulation
has been associated with progression of the malignancy [63],
highlighting CAV1 altered function depending on the organ
microenvironment studied.
Caveolin-1 single nucleotide polymorphism in renal
disease: Testa et al. found that there was a signi cant
independent and interaction association between CAV1
rs4730751 and eNOS rs1799983 genotype and increased carotid
arterial intima thickness (vascular hypertrophy) and cross-
sectional area across the common carotid artery (arterial
remodelling) [64]. It is not clear if these associations occur in
non-dialysis CKD or just a phenomenon seen in haemodialysis
patients. eNOS is bound to CAV1 requiring a calcium in ux for
its release from CAV1 in order to become activated.
Importantly, as CKD becomes more advanced the dominant
vascular lesion is arteriosclerosis and associated vascular
stiffness, rather than atheromatous disease as seen in the
general population [65]. Aortic pulse wave velocity (aPWV) is the
gold standard method for measuring arterial stiffness and has
been consistently associated with all-cause and cardiovascular
mortality in multiple conditions including CKD [66,67].
Independent of known clinical variables that in uence aPWV in
multivariate analysis, Chand et al. found that CAV1 rs4730751 CC
genotype is associated with lower arterial stiffness in patients
with early and late stage non-dialysis CKD [68]. In vascular
endothelium CAV1 interacts with eNOS such that reduced CAV1
increases eNOS activity which may have a deleterious effect on
endothelial health and arterial stiffness, due to “uncoupling”
of eNOS which leads to the generation of superoxide anion
radicals [69], in oxidative stress characteristic of CKD [70].
Similarly, caveolin-1 de cient aortic smooth muscle cells
have been shown to be pro-arteriosclerotic with increased
neointimal hyperplasia, cell proliferation and migration [71].
These observations may underlie the ndings of the current
study. Conversely, lower levels of CAV1 in macrophages are
associated with an anti-in ammatory phenotype, reduced
foam cell formation and therefore protection from atheroma
[57]. Interestingly, these contrasting functions of CAV1 in
endothelium (“anti-arteriosclerotic”) and macrophages
(“pro-atheromatous”) may consolidate the ndings of the
current study whereby CC genotype associates with a reduced
aPWV (“anti-arteriosclerotic”) and the ndings of Testa et al.
[64], who showed an association between CC genotype and
increased carotid arterial intima media thickness (a measure of
atheroma rather than arteriosclerosis [72].
Anti-neutrophil cytoplasmic antibody (ANCA) associated
vasculitis represent a group of primary autoimmune disorders
that are systemic, mainly involving small to medium sized
vessels. Despite success in improving patient life expectancy,
there remains high mortality at 5 years (up to 28%) and
signi cant morbidity associated with complications of the
disease and its treatment such as infection, cardiovascular
disease, malignancy and progression of kidney disease. These
complications mirror potential CAV1 effects, and indeed, the
CC genotype of CAV1 rs4739751 was associated with a better
outcome in this group of patients [73].
Donor CAV1 AA rs4739751 genotype of renal transplants are
associated with worsening renal allograft function, survival
as well as excess brosis upon histology in cohorts from
Birmingham [74], Belfast [74], and France [75].
012
Citation: Chand S (2018) Caveolin-1 in renal disease. Scientific J Genet Gene Ther 4(1): 007-014. DOI: http://dx.doi.org/10.17352/sjggt.000016
Conclusion
CAV1 pleotropic effects and ubiquitous distribution makes
it an attractive therapeutic target in many human diseases
in particular with patients in CKD, either to reduce related
morbidity and mortality, as a biomarker for identi cation of
high risk individuals, but also its direct manipulation in renal
brosis.
References
1. Coresh J, Selvin E, Stevens LA, Manzi J, Kusek JW, et al. (2007) Prevalence
of chronic kidney disease in the United States. Jama 298: 2038-2047. Link:
https://tinyurl.com/ybej3mkq
2. Kerr M, Bray B, Medcalf J, O’Donoghue DJ, Matthews B (2012) Estimating the
nancial cost of chronic kidney disease to the NHS in England. Nephrol Dial
Transplant 3: 73-80. Link: https://tinyurl.com/y7v7mlgb
3. Palade GE (1953) Fine Structure of Blood Capillaries. J Appl Phys 24: 1424-.
Link: https://tinyurl.com/yaxkzuv6
4. Yamada E (1955) The ne structure of the renal glomerulus of the mouse. J
Biophys Biochem Cytol 1: 551-566. Link: https://tinyurl.com/ybeffaoq
5. Cohen AW, Hnasko R, Schubert W, Lisanti MP (2004) Role of caveolae
and caveolins in health and disease. Physiol Rev 84: 1341-1379. Link:
https://tinyurl.com/yajh8g76
6. Fan JY, Carpentier JL, van Obberghen E, Grunfeld C, Gorden P, et al.
(1983) Morphological changes of the 3T3-L1 broblast plasma membrane
upon differentiation to the adipocyte form. J Cell Sci 61: 219-230. Link:
https://tinyurl.com/yad4r2rw
7. Scherer PE, Lewis RY, Volonte D, Engelman JA, Galbiati F, et al. (1997) Cell-
type and tissue-speci c expression of caveolin-2. Caveolins 1 and 2 co-
localize and form a stable hetero-oligomeric complex in vivo. J Biol Chem
272: 29337-2946. Link: https://tinyurl.com/yadnrdp4
8. Scheiffele P, Verkade P, Fra AM, Virta H, Simons K, et al. (1998) Caveolin-1
and -2 in the exocytic pathway of MDCK cells. J Cell Biol 140: 795-806. Link:
https://tinyurl.com/y7kjwsav
9. Tang Z, Scherer PE, Okamoto T, Song K, Chu C, et al. (1996) Molecular
cloning of caveolin-3, a novel member of the caveolin gene family
expressed predominantly in muscle. J Biol Chem 271: 2255-2261. Link:
https://tinyurl.com/yd3rlxlu
10. Rothberg KG, Heuser JE, Donzell WC, Ying YS, Glenney JR, et al. (1992)
Caveolin, a protein component of caveolae membrane coats. Cell 68: 673-
682. Link: https://tinyurl.com/yae2ao6h
11. Chidlow JH, Sessa WC (2010) Caveolae, caveolins, and cavins: complex
control of cellular signalling and in ammation. Cardiovasc Res 86: 219-225.
Link: https://tinyurl.com/y8eahgd5
12. Parton RG (2007) Simons K. The multiple faces of caveolae. Nat Rev Mol Cell
Biol 8: 185-194. Link: https://tinyurl.com/yc67m8az
13. Tanase C (2009) CAV1 (caveolin 1, caveolae protein, 22kDA). Atlas Genet
Cytogenet Oncol Haematol 13: 788-792. Link: https://tinyurl.com/y8cmcbxz
14. Engelman JA, Zhang XL, Lisanti MP (1999) Sequence and detailed
organization of the human caveolin-1 and -2 genes located near the D7S522
locus (7q31.1). Methylation of a CpG island in the 5’ promoter region of the
caveolin-1 gene in human breast cancer cell lines. FEBS Lett 448: 221-230.
Link: https://tinyurl.com/yd7xkukp
15. Engelman JA, Zhang X, Galbiati F, Volonte D, Sotgia F, et al. (1998) Molecular
genetics of the caveolin gene family: implications for human cancers,
diabetes, Alzheimer disease, and muscular dystrophy. Am J Hum Genet 63:
1578-1587. Link: https://tinyurl.com/y8pwqt7k
16. Schubert W, Frank PG, Razani B, Park DS, Chow CW, et al. (2001)
Caveolae-de cient endothelial cells show defects in the uptake and
transport of albumin in vivo. J Biol Chem 276: 48619-48622. Link:
https://tinyurl.com/yahzyjrn
17. Norkin LC, Anderson HA, Wolfrom SA, Oppenheim A (2002) Caveolar
endocytosis of simian virus 40 is followed by brefeldin A-sensitive transport
to the endoplasmic reticulum, where the virus disassembles. J Virol 76: 5156-
5166. Link: https://tinyurl.com/ycds7ejm
18. del Pozo MA, Balasubramanian N, Alderson NB, Kiosses WB, Grande-
Garcia A, et al. (2005) Phospho-caveolin-1 mediates integrin-regulated
membrane domain internalization. Nat Cell Biol 7: 901-908. Link:
https://tinyurl.com/yawwgn2w
19. Sharma DK, Brown JC, Choudhury A, Peterson TE, Holicky E, et al. (2004)
Selective stimulation of caveolar endocytosis by glycosphingolipids and
cholesterol. Mol Biol Cell 15: 3114-3122. Link: https://tinyurl.com/y75lbazb
20. Goligorsky MS, Li H, Brodsky S, Chen J (2002) Relationships between
caveolae and eNOS: everything in proximity and the proximity of everything.
Am J Physiol Renal Physiol 283: 1-10. Link: https://tinyurl.com/y7avdjob
21. Meng XM, Tang PM, Li J, Lan HY (2015) TGF-beta/Smad signaling in renal
brosis. Front Physiol 6: 82. Link: https://tinyurl.com/y8526uxx
22. Razani B, Zhang XL, Bitzer M, von Gersdorff G, Bottinger EP, et al. (2001)
Caveolin-1 regulates transforming growth factor (TGF)-beta/SMAD signaling
through an interaction with the TGF-beta type I receptor. J Biol Chem 276:
6727-6738. Link: https://tinyurl.com/y9nfvr55
23. Sigismund S, Argenzio E, Tosoni D, Cavallaro E, Polo S, et al. (2008)
Clathrin-mediated internalization is essential for sustained EGFR
signaling but dispensable for degradation. Dev Cell 15: 209-219. Link:
https://tinyurl.com/y9fcvxgx
24. Shi F, Sottile J (2008) Caveolin-1-dependent beta1 integrin endocytosis is
a critical regulator of bronectin turnover. J Cell Sci 121: 2360-2371. Link:
https://tinyurl.com/yd4sd8kt
25. Rizzo V, Morton C, DePaola N, Schnitzer JE, Davies PF (2003) Recruitment
of endothelial caveolae into mechanotransduction pathways by ow
conditioning in vitro. Am J Physiol Heart Circ Physiol 285: 1720-1729. Link:
https://tinyurl.com/ybqszn54
26. Yu J, Bergaya S, Murata T, Alp IF, Bauer MP, Lin MI, et al. (2006) Direct
evidence for the role of caveolin-1 and caveolae in mechanotransduction
and remodeling of blood vessels. J Clin Invest 116: 1284-1291. Link:
https://tinyurl.com/y8llvpw9
27. Sedding DG, Hermsen J, Seay U, Eickelberg O, Kummer W, et al. (2005)
Caveolin-1 facilitates mechanosensitive protein kinase B (Akt) signaling in
vitro and in vivo. Circ Res 96: 635-642. Link: https://tinyurl.com/y79uc7sm
28. Di Guglielmo GM, Le Roy C, Goodfellow AF, Wrana JL (2003) Distinct
endocytic pathways regulate TGF-beta receptor signalling and turnover. Nat
Cell Biol 5: 410-421. Link: https://tinyurl.com/y9frhfxx
29. Li Z, Wermuth PJ, Benn BS, Lisanti MP, Jimenez SA (2013) Caveolin-1
de ciency induces spontaneous endothelial-to-mesenchymal transition in
murine pulmonary endothelial cells in vitro. Am J Pathol 182: 325-331. Link:
https://tinyurl.com/yaky3n3n
30. Ito T, Williams JD, Fraser DJ, Phillips AO (2004) Hyaluronan regulates
transforming growth factor-beta1 receptor compartmentalization. J Biol
Chem 279: 25326-25332. Link: ‘https://tinyurl.com/yd4ffhh2
31. Zhang XL, Topley N, Ito T, Phillips A (2005) Interleukin-6 regulation of
transforming growth factor (TGF)-beta receptor compartmentalization and
turnover enhances TGF-beta1 signaling. J Biol Chem 280: 12239-12245. Link:
https://tinyurl.com/y77rencp
013
Citation: Chand S (2018) Caveolin-1 in renal disease. Scientific J Genet Gene Ther 4(1): 007-014. DOI: http://dx.doi.org/10.17352/sjggt.000016
32. Peng F, Wu D, Ingram AJ, Zhang B, Gao B, Krepinsky JC (2007) RhoA
activation in mesangial cells by mechanical strain depends on caveolae
and caveolin-1 interaction. J Am Soc Nephrol 18: 189-198. Link:
https://tinyurl.com/yb7jsvpq
33. Zhang Y, Peng F, Gao B, Ingram AJ, Krepinsky JC (2010) Mechanical
strain-induced RhoA activation requires NADPH oxidase-mediated
ROS generation in caveolae. Antioxid Redox Signal 13: 959-973. Link:
https://tinyurl.com/yaaqra2u
34. Bocanegra V, Manucha W, Pena MR, Cacciamani V, Valles PG (2010)
Caveolin-1 and Hsp70 interaction in microdissected proximal tubules from
spontaneously hypertensive rats as an effect of Losartan. J Hypertens 28:
143-155. Link: https://tinyurl.com/y9al6oxd
35. Nishiyama A, Hitomi H (2010) Role of caveolin and heat shock protein 70
interaction in the antioxidative effects of an angiotensin II type 1 receptor
blocker in spontaneously hypertensive rats proximal tubules. J Hypertens
28(1): 9-12. Link: https://tinyurl.com/ybk98ndv
36. Trujillo J, Ramirez V, Perez J, Torre-Villalvazo I, Torres N, et al. (2005) Renal
protection by a soy diet in obese Zucker rats is associated with restoration
of nitric oxide generation. Am J Physiol Renal Physiol 288: 108-116. Link:
https://tinyurl.com/ycj8weq4
37. Valles PG, Manucha W, Carrizo L, Vega Perugorria J, Seltzer A, et al. (2007)
Renal caveolin-1 expression in children with unilateral ureteropelvic junction
obstruction. Pediatr Nephrol 22: 237-248. Link: https://tinyurl.com/y9lmuntd
38. Moriyama T, Tsuruta Y, Shimizu A, Itabashi M, Takei T, et al. (2011) The
signi cance of caveolae in the glomeruli in glomerular disease. J Clin Pathol
64: 504-509. Link: https://tinyurl.com/ydxytkd7
39. Guan TH, Chen G, Gao B, Janssen MR, Uttarwar L, et al. (2013) Caveolin-1
de ciency protects against mesangial matrix expansion in a mouse
model of type 1 diabetic nephropathy. Diabetologia 56: 2068-2077. Link:
https://tinyurl.com/ydhhcpvw
40. Wu T, Zhang B, Ye F, Xiao Z (2013) A potential role for caveolin-1 in
VEGF-induced bronectin upregulation in mesangial cells: involvement
of VEGFR2 and Src. Am J Physiol Renal Physiol 304: 820-830. Link:
https://tinyurl.com/yc426rze
41. Herrera GA, Turbat-Herrera EA, Teng J (2016) Animal Models of Light
Chain Deposition Disease Provide a Better Understanding of Nodular
Glomerulosclerosis. Nephron. Link: https://tinyurl.com/ybggqt3e
42. Yamamoto I, Horita S, Takahashi T, Kobayashi A, Toki D, et al. (2008)
Caveolin-1 expression is a distinct feature of chronic rejection-
induced transplant capillaropathy. Am J Transplant 8: 2627-2635. Link:
https://tinyurl.com/y93ff8wy
43. Pontrelli P, Ursi M, Ranieri E, Capobianco C, Schena FP, et al. (2006) CD40L
proin ammatory and pro brotic effects on proximal tubular epithelial
cells: role of NF-kappaB and lyn. J Am Soc Nephrol 17: 627-636. Link:
https://tinyurl.com/y8xpnv6m
44. Park HC, Yasuda K, Ratliff B, Stoessel A, Sharkovska Y, et al. (2010)
Postobstructive regeneration of kidney is derailed when surge in renal stem
cells during course of unilateral ureteral obstruction is halted. Am J Physiol
Renal Physiol 298: 357-364. Link: https://tinyurl.com/ybfmdbrg
45. Chand S, Hazeldine J, Smith SW, Borrows R (2018) Genetic deletion of the
lipid raft protein caveolin-1 leads to worsening renal brosis. J Clin Nephrol
Ren Care 4: 037. Link: https://tinyurl.com/y9we8suq
46. Tourkina E, Richard M, Oates J, Hofbauer A, Bonner M, et al. (2010) Caveolin-1
regulates leucocyte behaviour in brotic lung disease. Ann Rheum Dis 69:
1220-1226. Link: https://tinyurl.com/y9xe3z43
47. Wang XM, Zhang Y, Kim HP, Zhou Z, Feghali-Bostwick CA, et al. (2006)
Caveolin-1: a critical regulator of lung brosis in idiopathic pulmonary
brosis. J Exp Med 203: 2895-2906. Link: https://tinyurl.com/yaub5tm8
48. Del Galdo F, Sotgia F, de Almeida CJ, Jasmin JF, Musick M, et al. (2008)
Decreased expression of caveolin 1 in patients with systemic sclerosis:
crucial role in the pathogenesis of tissue brosis. Arthritis Rheum 58: 2854-
2865. Link: https://tinyurl.com/ycymo8wg
49. Miyasato SK, Loe er J, Shohet R, Zhang J, Lindsey M, et al. (2011) Caveolin-1
modulates TGF-beta1 signaling in cardiac remodeling. Matrix Biol 30: 318-
329. Link: https://tinyurl.com/ycmmypq7
50. Zhang GY, Yu Q, Cheng T, Liao T, Nie CL, et al. (2011) Role of caveolin-1 in
the pathogenesis of tissue brosis by keloid-derived broblasts in vitro. Br J
Dermatol 164: 623-627. Link: https://tinyurl.com/y9x64v6r
51. Tsai TH, Chen SF, Huang TY, Tzeng CF, Chiang AS, et al. (2011) Impaired Cd14
and Cd36 expression, bacterial clearance, and Toll-like receptor 4-Myd88
signaling in caveolin-1-deleted macrophages and mice. Shock 35: 92-99.
Link: https://tinyurl.com/yanf9ym3
52. Gadjeva M, Paradis-Bleau C, Priebe GP, Fichorova R, Pier GB (2010) Caveolin-1
modi es the immunity to Pseudomonas aeruginosa. J Immunol 184: 296-
302. Link: https://tinyurl.com/y9fneylx
53. Guo Q, Shen N, Yuan K, Li J, Wu H, et al. (2012) Caveolin-1 plays a critical role
in host immunity against Klebsiella pneumoniae by regulating STAT5 and Akt
activity. Eur J Immunol 42: 1500-1511. Link: https://tinyurl.com/y8us3qaa
54. Zeitlin PL (2009) Pseudomonas aeruginosa: can studies in engineered cells
tell us why isit such a problem in people with cystic brosis? Focus on “Cystic
brosis transmembrane conductance regulator and caveolin-1 regulate
epithelial cell internalization of Pseudomonas aeruginosa”. Am J Physiol Cell
Physiol 297: 235-237. Link: https://tinyurl.com/y757gvhx
55. Sharon-Friling R, Shenk T (2014) Human cytomegalovirus pUL37x1-
induced calcium ux activates PKCalpha, inducing altered cell shape and
accumulation of cytoplasmic vesicles. Proc Natl Acad Sci U S A 111: 1140-
1148. Link: https://tinyurl.com/ycz69ch9
56. Moriyama T, Marquez JP, Wakatsuki T, Sorokin A (2007) Caveolar endocytosis
is critical for BK virus infection of human renal proximal tubular epithelial
cells. J Virol 81: 8552-8562. Link: https://tinyurl.com/y9m3ayeo
57. Fu Y, Moore XL, Lee MK, Fernandez-Rojo MA, Parat MO, et al. (2012) Caveolin-1
Plays a Critical Role in the Differentiation of Monocytes into Macrophages.
Arterioscler Thromb Vasc Biol. Link: https://tinyurl.com/ydhvl9db
58. Schwencke C, Schmeisser A, Walter C, Wachter R, Pannach S, et al. (2005)
Decreased caveolin-1 in atheroma: loss of antiproliferative control of vascular
smooth muscle cells in atherosclerosis. Cardiovasc Res 68: 128-315. Link:
https://tinyurl.com/ydggtltn
59. Zhou X, He P (2010) Endothelial [Ca2+]i and caveolin-1 antagonistically
regulate eNOS activity and microvessel permeability in rat venules.
Cardiovasc Res 87: 340-347. Link: https://tinyurl.com/y8yymcbs
60. Schwencke C, Braun-Dullaeus RC, Wunderlich C, Strasser RH (2006) Caveolae
and caveolin in transmembrane signaling: Implications for human disease.
Cardiovasc Res 70: 42-49. Link: https://tinyurl.com/yb5 4xr
61. Williams TM, Lisanti MP (2005) Caveolin-1 in oncogenic transformation,
cancer, and metastasis. Am J Physiol Cell Physiol 288: 494-506. Link:
https://tinyurl.com/y8vx6zso
62. Sotgia F, Martinez-Outschoorn UE, Howell A, Pestell RG, Pavlides S, et al.
(2012) Caveolin-1 and cancer metabolism in the tumor microenvironment:
markers, models, and mechanisms. Annu Rev Pathol 7: 423-467. Link:
https://tinyurl.com/y96lq4xh
63. Mathieu R, Klatte T, Lucca I, Mbeutcha A, Seitz C, et al. (2015) Prognostic
value of Caveolin-1 in patients treated with radical prostatectomy: a
multicentric validation study. BJU Int. Link: https://tinyurl.com/ybucyl8w
64. Testa A, Spoto B, Sanguedolce MC, Parlongo RM, Pisano A, et al. (2012) eNOS
and caveolin-1 gene polymorphisms interaction and intima media thickness:
014
Citation: Chand S (2018) Caveolin-1 in renal disease. Scientific J Genet Gene Ther 4(1): 007-014. DOI: http://dx.doi.org/10.17352/sjggt.000016
Copyright: © 2018 Chand S. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use,
distribution, and r eproduction in any medium, provided the original author and source are credited.
a proof of concept study in ESRD patients. Am J Hypertens 25: 103-108. Link:
https://tinyurl.com/yamms5sw
65. Chue CD, Townend JN, Steeds RP, Ferro CJ (2010) Arterial stiffness in
chronic kidney disease: causes and consequences. Heart 96: 817-823. Link:
https://tinyurl.com/ybkg7g3a
66. Vlachopoulos C, Aznaouridis K, Stefanadis C (2010) Prediction of
cardiovascular events and all-cause mortality with arterial stiffness: a
systematic review and meta-analysis. Journal of the American College of
Cardiology 55: 1318-1327.Link: https://tinyurl.com/yc36btk2
67. Baumann M, Wassertheurer S, Suttmann Y, Burkhardt K, Heemann U (2014)
Aortic pulse wave velocity predicts mortality in chronic kidney disease stages
2-4. J Hypertens 32: 899-903. Link: https://tinyurl.com/yacj2dqy
68. Chand S, Edwards NC, Chue CD, Jesky M, Stringer S, et al. (2015) Caveolin-1
single-nucleotide polymorphism and arterial stiffness in non-dialysis chronic
kidney disease. Nephrol Dial Transplant. Link: https://tinyurl.com/ycg66a8p
69. Tang L, Wang H, Ziolo MT (2014) Targeting NOS as a therapeutic approach
for heart failure. Pharmacol Ther. Link: https://tinyurl.com/yaxkjcwm
70. Massy ZA, Stenvinkel P, Drueke TB (2009) The role of oxidative
stress in chronic kidney disease. Semin Dial 22: 405-408. Link:
https://tinyurl.com/ycdrfc4x
71. Hassan GS, Williams TM, Frank PG, Lisanti MP (2006) Caveolin-1-de cient
aortic smooth muscle cells show cell autonomous abnormalities in
proliferation, migration, and endothelin-based signal transduction. Am J
Physiol Heart Circ Physiol 290: 2393-2401. Link: https://tinyurl.com/y6uljzz3
72. Matsushita K, Sang Y, Ballew SH, Shlipak M, Katz R, et al. (2015) Subclinical
atherosclerosis measures for cardiovascular prediction in CKD. J Am Soc
Nephrol 26: 439-447. Link: https://tinyurl.com/ycjyt3lu
73. Chand S, Holle JU, Hilhorst M, Simmonds MJ, Smith S, et al. (2013) Caveolin-1
single nucleotide polymorphism in antineutrophil cytoplasmic antibody
associated vasculitis. PLoS One 8: 69022. Link: https://tinyurl.com/y8rfzqxl
74. Moore J, McKnight AJ, Dohler B, Simmonds MJ, Courtney AE, et al. (2012)
Donor ABCB1 variant associates with increased risk for kidney allograft
failure. J Am Soc Nephrol 23: 1891-1899. Link: https://tinyurl.com/yaylsyj3
75. Van der Hauwaert C, Savary G, Pincon C, Gnemmi V, Noel C, et al. (2015)
Donor caveolin 1 (CAV1) genetic polymorphism in uences graft function
after renal transplantation. Fibrogenesis Tissue Repair 8: 8. Link:
https://tinyurl.com/yaylsyj3
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Background Renal disease is a major global public health issue. Renal interstitial fibrosis is the characteristic histopathological finding in all progressive renal disease. Caveolin-1 is the essential structural protein for lipid rafts called caveolae that are ubiquitously distributed among fibroblasts, endothelial and epithelial cells. Caveolin-1 acts as an intracellular signalling pathway chaperone in fibrotic disease. Presently, caveolin-1 expression is associated with more severe renal disease in human and previous murine studies. In non-renal fibrosis, caveolin-1 protects against fibrosis. The purpose of this study was to investigate if caveolin-1 knockout led to an increased fibrotic phenotype using the unilateral ureteric obstruction model of renal fibrosis. Methods Using 2 time-points of the unilateral ureteric obstruction model, wild-type and caveolin-1 knockout mouse kidneys were analysed for caveolin-1 expression and markers of fibrosis using histology, Gomori staining, real-time quantified polymerase chain reaction, Western blotting and confocal microscopy. Results Confocal microscopy shows caveolin-1 staining mainly in glomerulus, lining of tubules as well as the vasculature. There was increased caveolin-1 expression the longer the unilateral obstruction occurred as well as in the contralateral compensating non-obstructed kidney. Caveolin-1 knockout had less fibrosis at day 3 histologically but more at day 14 as compared to wild-type. There were significantly more F4/80 positive staining cells at day 3 and day 14 in the wild-type injured kidney as compared to the caveolin-1 knockout mouse. Conclusion Caveolin-1 knockout leads to a worse fibrosis upon unilateral ureteric obstruction. Caveolin-1 expression manipulation timing remains to be elucidated in reducing renal fibrosis.
Article
Full-text available
The obese Zucker rat is a valuable model for studying kidney disease associated with obesity and diabetes. Previous studies have shown that substitution of animal protein with soy ameliorates the progression of renal disease. To explore the participation of nitric oxide (NO) and caveolin-1 in this protective effect, we evaluated proteinuria, creatinine clearance, renal structural lesions, nitrites and nitrates urinary excretion (UNO2/NO3V), and mRNA and protein levels of neuronal NO synthase (nNOS), endothelial NOS (eNOS), and caveolin-1 in lean and fatty Zucker rats fed with 20% casein or soy protein diet. After 160 days of feeding with casein, fatty Zucker rats developed renal insufficiency, progressive proteinuria, and renal structural lesions; these alterations were associated with an important fall of UNO2/NO3V, changes in nNOS and eNOS mRNA levels, together with increased amount of eNOS and caveolin-1 present in plasma membrane proteins of the kidney. In fatty Zucker rats fed with soy, we observed that soy diet improved renal function, UNO2/NO3V, and proteinuria and reduced glomerulosclerosis, tubular dilation, intersticial fibrosis, and extracapilar proliferation. Renal protection was associated with reduction of caveolin-1 and eNOS in renal plasma membrane proteins. In conclusion, our results suggest that renal protective effect of soy protein appears to be mediated by improvement of NO generation and pointed out to caveolin-1 overexpression as a potential pathophysiological mechanism in renal disease.
Article
Full-text available
Background Arteriosclerosis is an independent predictor of increased cardiovascular mortality in chronic kidney disease (CKD). Histologically it is characterized by hypertrophy and fibrosis of the arterial media wall leading to increased arterial stiffness and end-organ damage. Caveolin-1 acts as an intracellular signalling pathway chaperone in human fibrotic and vascular diseases. The purpose of this study was to assess the association between caveolin-1 (CAV1) single-nucleotide polymorphism (SNP) rs4730751 and arterial stiffness as measured by arterial pulse wave velocity (PWV) in an early-stage CKD cohort and in a cohort with more severe CKD. Methods Two prospectively maintained patient cohorts with non-dialysis CKD were studied: 144 patients in the Chronic Renal Impairment in Birmingham (CRIB) cohort and 147 patients in the Renal Impairment in Secondary Care (RIISC) cohort, with matched exclusion criteria and DNA sampling availability. At entry to each cohort database, each patient's initial arterial PWV was measured, as well as their anthropomorphic and biochemical data. CAV1 rs4730751 SNP genotyping was performed using Taqman technology. Results The CAV1 rs4730751 SNP CC genotype was associated with lower arterial PWV in both CRIB early stage CKD patients [8.1 versus 8.6 m/s; coefficient −0.780 (−1.412, −0.149); P = 0.016] and RIISC more advanced stage CKD patients [8.7 versus 9.4 m/s; coefficient −0.695 (−1.288, −0.102); P = 0.022]; these relationships held following adjustment for other important confounders. Conclusions This replicated study suggests potential utility of the studied CAV1 SNP as a genetic biomarker in CKD and a role for CAV1 in the development of arteriosclerosis in this setting. Further studies are warranted to further explore the basic science driving these clinical observations.
Article
Full-text available
We have studied the biosynthesis and transport of the endogenous caveolins in MDCK cells. We show that in addition to homooligomers of caveolin-1, heterooligomeric complexes of caveolin-1 and -2 are formed in the ER. The oligomers become larger, increasingly detergent insoluble, and phosphorylated on caveolin-2 during transport to the cell surface. In the TGN caveolin-1/-2 heterooligomers are sorted into basolateral vesicles, whereas larger caveolin-1 homooligomers are targeted to the apical side. Caveolin-1 is present on both the apical and basolateral plasma membrane, whereas caveolin-2 is enriched on the basolateral surface where caveolae are present. This suggests that caveolin-1 and -2 heterooligomers are involved in caveolar biogenesis in the basolateral plasma membrane. Anti–caveolin-1 antibodies inhibit the apical delivery of influenza virus hemagglutinin without affecting basolateral transport of vesicular stomatitis virus G protein. Thus, we suggest that caveolin-1 homooligomers play a role in apical transport.
Article
Full-text available
Background Identification of the culprit genes underlying multifactorial diseases is one of the most important current challenges of molecular genetics. While recent advances in genomics research have accelerated the discovery of susceptibility genes, much remains to be learned about the functions of disease-associated genetic variants. Recently, Moore and co-workers identified, in the donor genome, an association between a common genetic variant (rs4730751) in the gene encoding caveolin-1 (CAV1), a major structural component of caveolae, and long-term allograft survival. Methods Four hundred seventy-five renal recipients consecutively transplanted were included in this study. Donor genomic DNA was extracted and used to genotype CAV1 rs4730751 Single Nucleotide Polymorphism. Results Patients receiving a graft carrying CAV1 rs4730751 AA genotype displayed a significant decrease in estimated glomerular filtration rate and a significant increase in serum creatinine in both univariate and multivariate analyzes. Moreover, patients receiving a graft with CAV1 AA genotype significantly developed more interstitial fibrosis lesions on systematic biopsies performed 3 months post-transplantation. Conclusions Genotyping of CAV1 may be relevant to identify patients at risk of adverse renal transplant outcome.
Article
Full-text available
TGF-β (transforming growth factor-β) is well identified as a central mediator in renal fibrosis. TGF-β initiates canonical and non-canonical pathways to exert multiple biological effects. Among them, Smad signaling is recognized as a major pathway of TGF-β signaling in progressive renal fibrosis. During fibrogenesis, Smad3 is highly activated, which is associated with the down-regulation of an inhibitory Smad7 via an ubiquitin E3-ligases-dependent degradation mechanism. The equilibrium shift between Smad3 and Smad7 leads to accumulation and activation of myofibroblasts, overproduction of ECM (extracellular matrix), and reduction in ECM degradation in the diseased kidney. Therefore, overexpression of Smad7 has been shown to be a therapeutic agent for renal fibrosis in various models of kidney diseases. In contrast, another downstream effecter of TGF-β/Smad signaling pathway, Smad2, exerts its renal protective role by counter-regulating the Smad3. Furthermore, recent studies demonstrated that Smad3 mediates renal fibrosis by down-regulating miR-29 and miR-200 but up-regulating miR-21 and miR-192. Thus, overexpression of miR-29 and miR-200 or down-regulation of miR-21 and miR-192 is capable of attenuating Smad3-mediated renal fibrosis in various mouse models of chronic kidney diseases (CKD). Taken together, TGF-β/Smad signaling plays an important role in renal fibrosis. Targeting TGF-β/Smad3 signaling may represent a specific and effective therapy for CKD associated with renal fibrosis.
Article
Full-text available
Whether inclusion of the coronary artery calcium score improves cardiovascular risk prediction in individuals with CKD, a population with unique calcium-phosphate homeostasis, is unknown. Among 6553 participants ages 45-84 years without prior cardiovascular disease in the Multi-Ethnic Study of Atherosclerosis, coronary artery calcium score was assessed for cardiovascular risk prediction beyond the Framingham predictors in those with (n=1284) and without CKD and contrasted with carotid intima-media thickness and ankle-brachial index (two other measures of subclinical atherosclerosis). During a median follow-up of 8.4 years, 650 cardiovascular events (coronary heart disease, stroke, heart failure, and peripheral artery disease) occurred (236 events in subjects with CKD). In Cox proportional hazards models adjusted for Framingham predictors, each subclinical measure was independently associated with cardiovascular outcomes, with larger adjusted hazard ratios (HRs; per 1 SD) for coronary artery calcium score than carotid intima-media thickness or ankle-brachial index in subjects without and with CKD (HR, 1.69; 95% confidence interval [95% CI], 1.45 to 1.97 versus HR, 1.12; 95% CI, 1.00 to 1.25 and HR, 1.20; 95% CI, 1.08 to 1.32, respectively). Compared with inclusion of carotid intima-media thickness or ankle-brachial index, inclusion of the coronary artery calcium score led to greater increases in C statistic for predicting cardiovascular disease and net reclassification improvement. Coronary artery calcium score performed best for the prediction of coronary heart disease and heart failure, regardless of CKD status. In conclusion, each measure improved cardiovascular risk prediction in subjects with CKD, with the greatest improvement observed with coronary artery calcium score.
Article
Background: Light chain deposition disease (LCDD) is a model of glomerulosclerosis. The mature lesion of LCDD mimics nodular glomerulosclerosis in diabetic nephropathy. The pathogenetic mechanisms involved are similar in both disorders, though the causative factors are entirely different. This fact highlights the generic response of the mesangium to varied stimuli. In-vitro work has provided much insight into the pathogenesis of glomerulosclerosis in LCDD where the mesangium is the main target for initiation and progression of the disease. The lack of animal models has prevented the development of further therapeutic approaches to be tested in platforms such as ex-vivo and in-vivo preparing the way for human studies. Methods: Light chains (LCs) obtained from the urine of patients with renal biopsy proven LCDD were delivered to glomeruli using ex-vivo and in-vivo approaches to address whether in-vitro information could be validated in-vivo. Selected in-vitro studies were conducted to address specific issues dealing with mesangial cell (MC) differentiation and composition of extracellular matrix to add additional data to the existing vast literature. Using light, electron and scanning microscopy together with immunohistochemistry and ultrastructural immunolabeling, MCs incubated in Matrigel with LCDD LCs, as well as delivery of such LCs by perfusion via renal artery (ex-vivo) and penile dorsal vein (in-vivo) to the kidneys, validation of pathogenetic pathways previously suggested in in-vitro experiments were tested and confirmed. Results: The animal models described in this manuscript provide validation for the in-vitro data that have been previously published and expand our appreciation of the important role that caveolin-1 plays in signaling events essential for the downstream sequence of events that eventually leads to the pathological alterations centered in the mesangium characterized by an increase in matrix production and formation of mesangial nodules. Conclusions: The same findings observed in renal biopsies of patients with LCDD (mesangial expansion with increased matrix) were documented in the ex-vivo and in-vivo platforms. In-vivo understanding of the pathogenesis of mesangial glomerulosclerosis, as accomplished in the reported research, is crucial for the design of novel therapeutic approaches to treat a number of glomerulopathies with similar pathogenetic mechanisms. Inhibiting interactions between glomerulopathic LCs and MCs or interrupting the protein production/secretion pathways are potentially effective therapeutic maneuvers. The results obtained with caveolin-1 knockout mice emphasized the importance of caveolin-1 in signaling events essential to effect downstream mesangial alterations.
Article
To validate Caveolin-1 as an independent prognostic marker of biochemical recurrence (BCR) in a large multi-institutional cohort of patients treated with radical prostatectomy (RP). Caveolin-1 expression was evaluated by immunochemistry on a tissue microarray from 3117 patients treated with RP for prostate cancer (PCa) at five institutions. Univariable and multivariable Cox proportional hazards regression models assessed the association of Caveolin-1 status with BCR. Harrell's C-index quantified prognostic accuracy (PA). Overexpression of Caveolin-1 was observed in 644 (20.6%) patients and was associated with higher pathological Gleason sum (p=0.002) and lymph node metastases (p=0.05). Within a median follow-up of 38 months (IQR 21-66), 617 (19.8%) patients experienced BCR. Patients with overexpression of Caveolin-1 had worse BCR free survival compared to patients with normal expression (log rank test, p=0.004). Caveolin-1 was an independent predictor of BCR in multivariable analyses that adjusted for the effects of standard clinicopathologic features (HR=1.21, p=0.037). Addition of Caveolin-1 in a model for prediction of BCR based on these standard prognosticators did not significantly improve predictive accuracy of the model. In subgroup analyses, Caveolin-1 was associated with BCR in patients with favorable pathologic features (pT2pN0 and Gleason score = 6) (p=0.021). We confirmed that the overexpression of Caveolin-1 is associated with adverse pathologic features in PCa and independently predicts BCR after RP, especially in patients with favorable pathologic features. However, it did not add prognostically relevant information to established predictors of BCR, limiting its use in clinical practice. This article is protected by copyright. All rights reserved. This article is protected by copyright. All rights reserved.