ArticlePDF Available

Cardiac SNARE Expression in Health and Disease

Frontiers
Frontiers in Endocrinology
Authors:

Abstract and Figures

SNARE proteins are integral to intracellular vesicular trafficking, which in turn is the process underlying the regulated expression of substrate transporters such as the glucose transporter GLUT4 at the cell surface of insulin target tissues. Impaired insulin stimulated GLUT4 trafficking is associated with reduced cardiac function in many disease states, most notably diabetes. Despite this, our understanding of the expression and regulation of SNARE proteins in cardiac tissue and how these may change in diabetes is limited. Here we characterize the array of SNARE proteins expressed in cardiac tissue, and quantify the levels of expression of VAMP2, SNAP23, and Syntaxin4—key proteins involved in insulin-stimulated GLUT4 translocation. We examined SNARE protein levels in cardiac tissue from two rodent models of insulin resistance, db/db mice and high-fat fed mice, and show alterations in patterns of expression are evident. Such changes may have implications for cardiac function.
Content may be subject to copyright.
ORIGINAL RESEARCH
published: 19 December 2019
doi: 10.3389/fendo.2019.00881
Frontiers in Endocrinology | www.frontiersin.org 1December 2019 | Volume 10 | Article 881
Edited by:
Debbie C. Thurmond,
Beckman Research Institute,
United States
Reviewed by:
Amira Klip,
Sick Kids Research Institute, Canada
Jonathan Bogan,
Yale University, United States
*Correspondence:
Gwyn W. Gould
gwyn.gould@strath.ac.uk
Present address:
Peter R. T. Bowman and
Gwyn W. Gould,
Strathclyde Institute of Pharmacy and
Biomedical Sciences, Glasgow,
United Kingdom
Specialty section:
This article was submitted to
Molecular and Structural
Endocrinology,
a section of the journal
Frontiers in Endocrinology
Received: 25 September 2019
Accepted: 03 December 2019
Published: 19 December 2019
Citation:
Bowman PRT, Smith GL and
Gould GW (2019) Cardiac SNARE
Expression in Health and Disease.
Front. Endocrinol. 10:881.
doi: 10.3389/fendo.2019.00881
Cardiac SNARE Expression in Health
and Disease
Peter R. T. Bowman 1†, Godfrey L. Smith 2and Gwyn W. Gould 1
*
1Henry Wellcome Laboratory of Cell Biology, College of Medical, Veterinary and Life Sciences, Institute of Molecular Cell and
Systems Biology, University of Glasgow, Glasgow, United Kingdom, 2College of Medical, Veterinary and Life Sciences,
Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
SNARE proteins are integral to intracellular vesicular trafficking, which in turn is the
process underlying the regulated expression of substrate transporters such as the
glucose transporter GLUT4 at the cell surface of insulin target tissues. Impaired insulin
stimulated GLUT4 trafficking is associated with reduced cardiac function in many disease
states, most notably diabetes. Despite this, our understanding of the expression and
regulation of SNARE proteins in cardiac tissue and how these may change in diabetes is
limited. Here we characterize the array of SNARE proteins expressed in cardiac tissue,
and quantify the levels of expression of VAMP2, SNAP23, and Syntaxin4—key proteins
involved in insulin-stimulated GLUT4 translocation. We examined SNARE protein levels
in cardiac tissue from two rodent models of insulin resistance, db/db mice and high-fat
fed mice, and show alterations in patterns of expression are evident. Such changes may
have implications for cardiac function.
Keywords: diabetes, cardiomyopathy, SNARE proteins, insulin resistance, GLUT4
INTRODUCTION
Effective regulation of metabolism is essential in all cell types in order to ensure that ATP generation
requirements are met. This is particularly important within highly energetic organs such as the
heart, where the contractile action of cardiomyocytes must be continually fuelled in order to
maintain the pumping of approximately 5 liters of oxygen and nutrient rich blood into and around
the systemic circulation every minute. It is also vital that the heart exhibits metabolic flexibility in
order to adapt its contractile output in response to increased demands e.g., during exercise. Normal
cardiac metabolism is characterized by predominant use of fatty acids as a metabolic substrate, with
a relatively lower utilization of glucose (1). This is logical as fat is a more abundant and energy rich
fuel source, making it ideal for scenarios where sustained moderate levels of ATP are required.
However, several cardiac disease states are partly defined (and potentially caused) by deficits in
glucose uptake and metabolism.
The predominant type 2 diabetic phenotype is characterized by peripheral insulin resistance,
whereby insulin no longer effectively stimulates the uptake of glucose into muscle and adipose
tissue. The mechanism underlying this condition is multifactorial, but is strongly linked to
obesity (2). This physiological action of insulin is important in maintaining glycaemic control
post-prandially, and indeed diabetes is diagnosed by an elevation in fasting blood glucose (or
circulating glycated hemoglobin) above clinically defined thresholds. In a contracting working
heart preparation from a diabetic mouse model, radiolabelled substrates revealed an impairment
of insulin stimulated glycolysis and glucose oxidation and increased basal fatty acid utilization
(associated with reduced myocardial efficiency) and an impaired ability of insulin to reduce this (3).
Bowman et al. Cardiac SNARE Proteins
Additionally, increasing the rate of fatty acid delivery to control
hearts resulted in reduced glycolysis and glucose oxidation,
enhanced fatty acid utilization, and reduced cardiac performance
(3). This demonstrates the importance of insulin-stimulated
glucose uptake to fatty acid metabolism (and vice versa) and
therefore cardiac efficiency and performance and exemplifies
potential defects in these systems in diseases such as diabetes.
Cardiovascular disease has long been established to be a
leading cause of death in the diabetic population, in part
attributable to the high rate of vascular disease that occurs
on account of sustained excessive glucose (and fat) in the
bloodstream (4). However, there is also a direct pathological
effect of diabetes upon cardiac function, characterized by
initial diastolic dysfunction prior to structural remodeling and
progression to heart failure, termed diabetic cardiomyopathy (5
7). There is strong evidence that metabolic impairments such
as the intramyocellular accumulation of lipids in the heart and
associated cardiac insulin resistance may be critical early factors
in the progression of this condition (810). Most notably, in a
mouse model of diabetic cardiomyopathy, overexpression of the
insulin sensitive glucose transporter GLUT4 restored aberrant
cardiac metabolic and contractile function to values observed
in controls (11,12). Additionally, insulin resistance/glycaemic
control has also been demonstrated to be of prognostic
significance in human post-myocardial infarction patients (13
15), with an experimental rat model identifying the onset of
cardiac (not systemic) insulin resistance to be correlated with
adverse recovery/remodeling (16).
It has been established that general concepts derived from
other insulin sensitive cell types are applicable to the heart, for
example that GLUT4 is the functionally predominant glucose
transporter (17). Under basal conditions the majority of GLUT4
is not located at the cell surface but rather distributed between the
general endosomal recycling pathway and a depot of specialized
GLUT4 storage vesicles (GSVs) that can be rapidly mobilized
to the cell surface in response to activation of the insulin
receptor (18). Both the sorting of GLUT4 through different
intracellular compartments and the fusion of GSVs with the
plasma membrane (PM) requires the action of specific SNARE
proteins (19). In skeletal muscle and fat tissue, the SNARE
proteins that regulate GSV fusion with the PM are VAMP2,
SNAP23, and Syntaxin 4, whereas Syntaxin 6 and 16 mediate
sequestration of GLUT4 into pools of GSVs at the trans Golgi
network (2025).
Numerous studies have used a variety of techniques (including
knock down or expression of dominant mutant proteins)
to disrupt the function of SNARE proteins in the insulin
sensitive cell model 3T3-L1 adipocytes and demonstrated a clear
inhibitory effect upon insulin dependent GLUT4 trafficking and
glucose uptake, with the precise defect depending on the protein
targeted (2630). Furthermore, in vivo examination of mice over
or under-expressing Syntaxin 4 identified improved/impaired
skeletal muscle insulin stimulated glucose uptake and therefore
also whole body glucose tolerance, attributed to corresponding
effects on insulin stimulated GLUT4 translocation (31,32).
To underline this strong association between SNARE proteins,
GLUT4 trafficking, and glycaemic control, even manipulation of
the expression of ancillary proteins such as Munc18c and Doc2b
that regulate SNARE interactions has significant consequences
on GLUT4-PM integration and glucose uptake (33,34). Insulin-
stimulated GLUT4 translocation in peripheral tissues thus
exhibits a considerable degree of mechanistic overlap, with
studies emphasizing the role of Syntaxin 4 and SNAP23 in fat and
muscle cells, and transgenic mice clearly establishing that levels of
expression of these SNAREs correlate with whole-body glycaemic
control (31,32).
The involvement of SNAREs in multiple steps of GLUT4
trafficking make them an intriguing potential target in the
context of disease. There are many theories relating to insulin
resistance, such as inhibition of proximal insulin signaling via
either lipid mediated activation of protein kinase C (35,36) or
altered release of adipocytokines from expanded and inflamed
adipose tissue (37,38). However, there is also evidence from
rodent models correlating altered SNARE protein (Syntaxin4,
Syntaxin6, VAMP2, VAMP3, SNAP23, Munc18) expression or
localization with skeletal muscle and adipose insulin resistance
(31,32,3941). Additionally, in human type 2 diabetic patients
enhanced syntaxin 8 expression in adipose tissue was significantly
associated with reduced GLUT4 expression and impaired whole
body glucose tolerance (42). While it is not clear from these
studies whether alterations in SNARE protein levels are causal
or adaptive changes, initial studies from 2 independent insulin
resistant models indicate that targeting SNARE proteins may be
a viable strategy to restore insulin stimulated GLUT4 trafficking
and improve metabolic outcomes (4345).
Any attempt to investigate these initial observations in the
heart is constrained by very limited prior characterization of
SNARE protein expression in cardiomyocytes, let alone how
they may interact or be of functional importance. This is in
contrast to considerable investigation in adipose and skeletal
muscle. The only specific prior attempt to identify the expression
and involvement of SNAREs in cardiac GLUT4 trafficking was
performed in a mouse atrial cell line (HL-1) and restricted to
assessment of all VAMP (v-SNARE) isoforms (46). Expression
of VAMP2/3/4/5/7 (but not VAMP1 or 8) protein was detected,
and this was confirmed in lysate generated from mouse heart.
Targeted silencing of each isoform revealed a role for VAMP2
and VAMP5 in the insulin stimulated appearance of GLUT4-myc
at the PM. However, separate work with rodent cardiomyocytes
identified the expression of VAMP1/2/3/4/5/8 (but not VAMP7)
mRNA transcripts (47). Whilst this study did not investigate
the expression of all VAMPs beyond the use of RT-PCR, this
data highlights that even within this limited field contradictions
have emerged regarding the expression of VAMP1/7/8 in the
heart. It is possible that this is in part due to the use of isolated
cardiomyocytes (47). Whilst isolation of cells prior to analysis
reduces interference in the sample from other subpopulations of
cardiac cells, if maintained in culture dedifferentiation may start
to occur which could impact protein expression.
Therefore, the aim of this work was to characterize the
expression of a wide range of SNARE isoforms within primary
adult cardiac tissue. Furthermore, it was assessed if the expression
of these proteins (in addition to GLUT4) was altered in 2
different diabetic mouse models. This study is the first step
Frontiers in Endocrinology | www.frontiersin.org 2December 2019 | Volume 10 | Article 881
Bowman et al. Cardiac SNARE Proteins
FIGURE 1 | Quantification of Syntaxin 4 expression in the heart. Protein lysates were generated from 20-week old male mouse cardiac tissue and subjected to
SDS-PAGE and immunoblotting alongside purified recombinant Sx4. Lysates were incubated with antibodies probing for the expression of Syntaxin 4 and GAPDH
(loading control) as shown. Protein loaded refers to 10 or 20 µg of protein for the M1-M3 mouse cardiomyocyte samples or 0.5–1.5 ng of purified Syntaxin 4 protein
(lacking the transmembrane domain and thus migrating faster than endogenous, full length Syntaxin4), as indicated. M1-3 indicates biologically independent mouse
cardiac samples, where each sample is composed of lysate generated from 2 to 3 individual hearts. The approximate positions of molecular weight markers are
indicated. Borders indicate where images have been cropped for presentation purposes only. Data from a representative immunoblot is shown, quantified in Table 1.
TABLE 1 | Quantification of SNARE protein expression in primary mouse cardiac tissue.
Sample SNAP23 Copies/mg Syntaxin 4 Copies/mg VAMP2 Copies/mg
Mouse Heart 2.07 ×1012 (±5.66 ×1011) 7.26 ×1011 (±2.67 ×1010) 3.23 ×1011 (±3.56 ×1010 )
3T3-L1 Adipocytes 5.28 ×1012 1.72 ×1012 2.28 ×1012
Densitometry was used to quantify Syntaxin 4, SNAP23, and VAMP2 expression from known amounts of primary cardiac tissue and purified recombinant protein, within immunoblots
such as that depicted in Figure 1. Values for recombinant proteins were used to generate a standard curve that allowed estimation of the absolute amount of each protein in a given
amount of total cardiac lysate. This value is expressed in copies per mg of total lysate and is the mean (±S.E.M.) of at least 3 biologically independent replicate samples. Values from
3T3–L1 adipocytes are those reported in Hickson et al. (48).
toward uncovering the role of different SNAREs in insulin
stimulated cardiac GLUT4 trafficking and is of clinical relevance
due to the association of cardiac insulin resistance with diabetic
cardiomyopathy and myocardial infarction. SNAREs are also
important in both non-insulin stimulated (e.g., contraction
mediated) GLUT4 trafficking and the trafficking of the fatty
acid transporter CD36 (46). Therefore, this work is valuable
and foundational in the context of the regulation of cardiac
metabolism in general.
RESULTS AND DISCUSSION
Quantification of VAMP2, Syntaxin 4, and
SNAP23 in Cardiac Lysates
In adipocytes and muscle, the SNARE proteins associated with
GSV-PM fusion are VAMP2, Syntaxin 4, and SNAP23 (26,27,
30). We first sought to quantify expression of these SNAREs
in rodent cardiac lysates. Recombinant SNARE proteins were
expressed and purified from bacteria and used as standards in
quantitative immunoblotting to compare to the signal obtained
from mouse cardiac samples. An example of this technical
approach is shown in Figure 1 for quantification of Syntaxin
4 expression, with quantification of all 3 SNAREs reported in
Table 1. Comparative values displayed for 3T3-L1 adipocytes
were obtained from previously published work using a similar
approach (48). This technique was also performed with a
pooled lysate generated from cardiac tissue derived from 3
individual rat hearts. The mean values obtained closely matched
those of the mouse samples (1.45 ×1012 copies per mg
SNAP23; 6.82 ×1011 copies per mg Sx4; 3.05 ×1011 copies
per mg VAMP2).
In 3T3-L1 adipocytes there are approximately 2–3 fold more
SNAP23 molecules in comparison to VAMP2 and Syntaxin 4
(48). It is acknowledged that within this study our technical
approach will be associated with a margin of error, however
this phenomenon may not be specific to adipocytes as a
similar relative expression of key SNARE proteins is recorded
here from two sources of primary cardiomyocytes. The reason
underlying this observation requires further study but most
plausibly may reflect the involvement of SNAP23 in multiple
other fusion events relevant to other biological processes.
Additionally, this data provides one possible explanation as to
why 3T3-L1 adipocytes display a greater fold insulin response
compared to primary cardiomyocytes. Despite expressing less
GLUT4 (49), 3T3-L1 adipocytes express abundantly more of
each SNARE protein thereby generating a more favorable
ratio between the availability of transporters and fusion
machinery. However, it must also be acknowledged that the
heart tends to display an elevated basal rate of glucose
uptake in order to support contractile demands (50), which
would therefore reduce any fold insulin response. These data
also confirm that, like 3T3-L1 adipocytes, Syntaxin 4 and
Frontiers in Endocrinology | www.frontiersin.org 3December 2019 | Volume 10 | Article 881
Bowman et al. Cardiac SNARE Proteins
FIGURE 2 | Cardiac SNARE expression in db/db and db/m mice. Protein lysates were generated from db/db and control mouse hearts and subjected to SDS-PAGE
and immunoblotting, probing for the expression of numerous SNARE proteins and GLUT4. Representative immunoblots for selected proteins only are shown. A
representative Ponceau S staining of a membrane is displayed, which was used as a loading control. For each sample, 40 µg of protein lysate was loaded. Each
sample (1–6 on figure) is composed of lysate generated from a separate heart.
VAMP2 are expressed at broadly similar levels on a per
cell basis.
SNARE Expression in Diabetic Mouse
Models
Insulin resistance may have an important role in the development
of limitations in cardiac contractile performance in diabetic
individuals. Accordingly, given their involvement in multiple
steps of GLUT4 trafficking, SNARE proteins may be implicated
in this clinically relevant pathophysiological setting. It is
therefore necessary to characterize the array of SNARE trafficking
machinery present in primary cardiac tissue in order to probe
known functions of these proteins (in other cell types) in the
heart, and novel physiological roles or interactions specific to
cardiac function. It is challenging to obtain a significant amount
of human myocardial samples for biochemical investigation,
particularly from specific patient populations and appropriately
matched controls. In order to circumnavigate this issue, many
animal models of disease have been developed in the field of
cardiac physiology. In the context of diabetes one of the most
widely utilized and characterized models is the db/db mouse,
which has a leptin receptor mutation that results in obesity
and blood parameters mimicking the human diabetic state
(51). Importantly, this model not only displays systemic insulin
resistance, but also cardiomyocyte-specific insulin resistance
and reduced cardiac contractility (recorded at the level of the
whole heart) (52,53), making it an ideal tool for investigation
of DCM.
Within this study, cardiac segments from 6 db/db and control
(db/m) mice were lysed and probed for the expression of a
broad range of SNARE proteins and GLUT4 via immunoblotting.
A typical dataset is displayed in Figure 2. Consistent with
previous analysis of cardiac protein expression from this diabetic
mouse model (53,54), GLUT4 expression was found to be
significantly (P<0.05) reduced in db/db lysates. We observed
clear and consistent immunoblot signals for Sx2, 4, 5, 8,
and 16, SNAP23, 29 and 47, and VAMP2, 3, 4, 5, and 8
(Supplementary Table 1), but as shown for a subset in Figure 2,
Frontiers in Endocrinology | www.frontiersin.org 4December 2019 | Volume 10 | Article 881
Bowman et al. Cardiac SNARE Proteins
FIGURE 3 | Cardiac SNARE expression in HFD fed and control mice. Protein lysates were generated from HFD and control mouse hearts and subjected to
SDS-PAGE and immunoblotting, probing for the expression of SNARE proteins and GLUT4. Representative immunoblots for selected proteins are shown together
with Ponceau S staining, which was used as a loading control. Each sample is composed of lysates generated from 2 to 4 individual hearts.
no significant differences between db/m and db/db mice were
observed for any SNARE proteins. In contrast to Schwenk et al.
(46) but consistent with Peters et al. (47), we were unable to
detect VAMP7 in primary cardiac tissue yet obtained a clear
signal for VAMP8. However, in support of Schwenk et al. (46)
but in opposition to Peters et al. (47), we were unable to
detect VAMP1.
The db/db mouse is considered to be a good representation
of the established human diabetic phenotype. However,
there are legitimate questions regarding the translational
relevance of this model. Whilst there is almost certainly a
genetic component in many cases of type 2 diabetes (55),
more commonly lifestyle factors—e.g., food intake and
activity levels—that determine overall net energy balance
are recognized as key factors in diabetes prevalence. In
particular, obesity greatly increases the risk of an individual
developing diabetes (2). Therefore, high fat diet (HFD) based
interventions in rodents have been developed extensively.
This produces a less severe diabetic phenotype typically
analogous to a pre-diabetic state (56), however still captures
reduced cardiac contractility in the context of insulin resistance
(57). It is likely that the mechanisms underpinning complex
phenotypes such as insulin resistance and cardiomyopathy
may be multifactorial and vary with disease progression.
Therefore, we examined SNARE protein expression in a mouse
HFD model.
As can be observed in Figure 3 and Table 2, an almost
identical range of SNARE proteins were detected in this second
independent mouse cohort as observed in Figure 2. Due to
limitations in sample availability, lysates from 2 to 4 individual
hearts were pooled in order to generate each of the 4 samples
displayed, which reduced the statistical power of subsequent
analysis. With this limitation in mind, there was no impact of
the pro-diabetic phenotype upon GLUT4 expression. However,
statistically significant elevations in VAMP5 (78% increase) and
SNAP29 (68%) expression were detected. It appeared as though
VAMP2 expression may have been reduced in at least one
experimental group, however this effect was not found to be
statistically significant.
The samples used in Figures 2,3were obtained from sections
of frozen tissue. Therefore, measurements could not be obtained
that could confirm or refute that these lysates were from insulin
resistant hearts. However, we note that the db/db mouse is
a reliable and well characterized model and our observation
of reduced GLUT4 expression supports the disease phenotype
being evident in these samples. Importantly, published data from
the same control and HFD mice cohort confirms that samples
used here were from mice that displayed significant weight gain
Frontiers in Endocrinology | www.frontiersin.org 5December 2019 | Volume 10 | Article 881
Bowman et al. Cardiac SNARE Proteins
TABLE 2 | Quantification of SNARE protein expression in HFD primary cardiac lysates.
Protein Detected? Difference? Protein Detected? Difference?
SNAP23 Yes No Syntaxin 16 Yes No
SNAP29 Yes Yes, significantly (P=0.04)
increased in HFD lysates by 68%
GLUT4 Yes No
SNAP47 Yes No VAMP1 No N/A
Syntaxin 2 Yes No VAMP2 Yes 30% lower expression in HFD
lysates, difference was not
significant (P=0.33)
Syntaxin 3 Yes No VAMP3 Yes No
Syntaxin 4 Yes No VAMP4 Yes No
Syntaxin 5 Yes No VAMP5 Yes Yes, significantly (P=0.02)
increased in HFD lysates by 78%
Syntaxin 6 Yes No VAMP7 No N/A
Syntaxin 7 No N/A VAMP8 Yes No
Syntaxin 8 Yes No
Immunoblotting was used to detect the presence or absence of a range of proteins in cardiac lysates generated from HFD and CHOW control mice. Where a clear and reproducible
difference was observed between groups, densitometry was used to quantify expression relative to total protein (from Ponceau stained images) and an unpaired Student’s t-test was
used to assess statistical significance (N =2). The level of significance was set at P =0.05.
and a trend toward delayed systemic glucose clearance during
a glucose tolerance test compared to controls—indicative of
reduced insulin sensitivity (58).
We have shown a range of SNARE proteins are expressed
in primary cardiac samples. This exceeds the machinery we
may expect to be required for the regulated trafficking of
substrate transporters such as GLUT4 or CD36. Indeed, the
heart is known to secrete vasoactive natriuretic peptides in
order to regulate blood pressure and therefore cardiac loading,
and recent work has also highlighted the emerging role that
numerous cardiokines may have in modulating both cardiac
remodeling in response to sustained pathophysiological stress
through auto/paracrine signaling and also remote physiological
functions through endocrine actions (59). The secretion of
these signaling proteins will be reliant upon the action of
SNAREs. Our study will provide the platform necessary to
relate SNARE function to specific physiological actions, and
therefore ultimately investigate any potential role in disease.
Additionally, this study provides clarity where there had
previously been conflicting data regarding the expression of
VAMP1/7/8. Obtaining identical results for these proteins
from 2 independent mouse lines provides confidence in
these findings.
Interestingly, in the HFD model SNAP29 and VAMP5
expression were elevated relative to control samples. VAMP5
has previously been linked to GLUT4 trafficking, but only in a
cardiomyocyte cell line model (46). In contrast, the functional
role of SNAP29 is not clear, particularly in the heart. Given
that impaired GLUT4 trafficking underlies insulin resistance, it
is unclear why expression of a related protein would remain
elevated beyond any initial compensatory period. It is interesting
to note however that elevated SNARE protein levels were
observed in skeletal muscle of the ZDF rat model of diabetes,
which were reversed by treatment with thiazolidinediones (39).
Hence, there is a precedent for elevated SNARE expression in
insulin resistance. In contrast to the study of skeletal muscle
insulin resistance which saw elevations in Sx4 and VAMP2,
here we report elevated VAMP5 in cardiac tissue from HFD
mice. VAMP5 has also been implicated in the regulation of
contraction-mediated glucose uptake in cardiomyocytes (46)
and thus our observation may reflect a functional distinction
between skeletal muscles and cardiomyocytes. Elevated VAMP5
may change (or reflect) the contractile profile of the heart in the
early stages of diabetes prior to the onset of cardiomyopathy.
It might be speculated that enhanced contraction is used to
compensate for reduced insulin mediated glucose uptake at
certain times. Alternatively, the observed changes may relate
to the trafficking of another target, perhaps a fatty acid
transporter. Enhanced cardiac lipid content is one of several
factors in the onset of insulin resistance, but this can only
occur with increased trafficking of the relevant transporters to
the cell membrane. Further work will be required to answer
these questions and to define whether the changes observed
in cardiac SNARE proteins in disease models are causal or
adaptive responses.
CONCLUSION
This study has detailed, for the first time, the expression of a
wide range of SNARE proteins in primary cardiac samples. This
foundational work paves the way for future studies investigating
the interactions and roles of these proteins in several different
aspects of cardiac physiology, both under normal working
conditions and in disease.
MATERIALS AND METHODS
Primary Cardiac Samples
All primary cardiac tissues used during this study were kind
gifts from colleagues at the University of Glasgow; procedures
were undertaken in accordance with the United Kingdom
Animals (Scientific Procedures) Act of 1986 and conform
Frontiers in Endocrinology | www.frontiersin.org 6December 2019 | Volume 10 | Article 881
Bowman et al. Cardiac SNARE Proteins
TABLE 3 | Information regarding primary cardiac samples.
Sample Key details Gifted by
Db/db and db/m
mice
13–15-week-old male mice either hetero
or homozygous for a mutation in the leptin
receptor gene
Dr. Augusto
Montezano
High Fat Diet fed
and control mice
Approximately 20-week-old male mice fed
standard CHOW diet for 8 weeks
postnatally prior to switching to a diet
whereby 60% of calories were obtained
from fat for the next 12 weeks (or
maintained on CHOW as control)
Dr. Anna
White
to the Guide for the Care and Use of Laboratory Animals
published by the US National Institutes of Health and
approved by the Glasgow University Ethical Review Board.
In all cases tissues were received as sections of myocardium
that had been snap frozen either at 80C or in liquid
nitrogen. Key details pertaining to each sample are listed
in Table 3. All of the samples gifted were from male
animals, therefore it is acknowledged that there may be
sex specific differences in the reported findings that require
further investigation.
Generation of Lysates
The procedure for generation of cardiac lysates was the
same regardless of the origin of the tissue. First of all, a
large section of myocardium was excised from each heart.
These sections were generated in the same way by the same
experimenter, without prior separation into distinct anatomical
regions. The vast majority of myocardium is ventricular tissue,
and therefore any large section of cardiac tissue will almost
certainly originate predominantly from this area. Each section
was placed within a plastic 10 cm dish on ice in RIPA buffer
at a ratio of approximately 1-part tissue to 5 parts buffer.
The tissue was then manually diced with a sterile scalpel
blade into sections as small as possible, prior to further
mechanical lysis via a Dounce style tissue grinder. Samples were
placed on ice for a further 20 min, prior to repetition of this
mechanical lysis technique. The lysates were then centrifuged at
17,500 g for 15 min at 4C. The supernatant was then collected,
and the pellet was discarded. Prior to immunoblotting the
protein concentration of each sample was calculated via micro
BCA assay.
Generation and Quantification of Purified
Protein
Recombinant purified protein samples were generated as
described previously by Sadler et al. (60). First of all, plasmids
encoding sequences for Syntaxin 4, SNAP23, and VAMP2
tagged with glutathione S-transferase (GST) but in the case
of Sx4 and VAMP2 lacking their transmembrane domains
to increase expression were transformed into BL-21 E.coli
and subsequently amplified in serial cultures, using ampicillin
(100 µg/mL) resistance as a selection marker. Once in the optimal
growth range and volume, recombinant protein production was
induced via Isopropyl B-D-1-thiogalactopyranoside (0.5 mM)
incubation overnight at 22C. The following day cells were
lysed, and target proteins were extracted through incubation
of the lysate for 2 h with glutathione beads. Thereafter,
the beads were washed with PBS and then the samples
were eluted. Prior to use within this study, the GST tags
were cleaved from the proteins. Quantification of purified
protein concentration was performed by running samples
through SDS-PAGE against known amounts of BSA and
then performing Coomassie staining in order to visualize all
protein bands, as described by Sadler et al. (60). Gels were
scanned and then densitometry was performed in order to
generate a protein standard curve with BSA signals, which
was then used to estimate the concentration of each purified
SNARE sample.
SDS-PAGE and Immunoblotting
Samples were defrosted on ice, combined 1:1 with 2x Laemmli
sample buffer, and then heated to 65C for 10 min. Homemade
polyacrylamide gels of the appropriate percentage were cast
and then lysates were loaded in order to achieve the desired
(often equal) amount of protein across different samples. These
were then separated via gel electrophoresis and subsequently
subjected to immunoblotting as described previously by Sadler
et al. (60). Densitometry was performed via Image Studio Lite
in order to quantify the generated images. Either GAPDH
expression or Ponceau S staining was used in order to assess total
protein loading.
Antibodies
Anti-GLUT1 (#652) and anti-GLUT4 (#654) were from AbCam
(Cambridge, United Kingdom). Anti-GAPDH (#4300) was from
Ambion (Foster city, California, USA). Anti-VAMP1 (#104002),
anti-VAMP2 (#104202), anti-VAMP3 (#43080), anti-VAMP4
(#136002), anti-VAMP5 (#176003), anti-VAMP7 (#232003),
anti-VAMP8 (#104302), anti-SNAP23 (#111202), anti-SNAP29
(#111303), anti-SNAP47 (#111403), anti-syntaxin2 (#110022),
anti-syntaxin3 (#110032), anti-syntaxin4 (#110042), anti-
syntaxin5 (#110053), anti-syntaxin7 (#110072), anti-syntaxin8
(#110083), and anti-syntaxin16 (#110162) were from Synaptic
Systems (Goettingen, Germany). Anti-syntaxin6 (#610635)
was from BD Biosciences (Franklin lakes, New Jersey, USA).
Fluorescent secondary antibodies were from LI-COR Biosciences
(Lincoln, Nebraska, USA).
Statistical Analysis
Statistical testing was performed with GraphPad Prism 7. Where
appropriate, this consisted of an unpaired t-test, assessing the
difference in protein expression between one of the experimental
groups and relevant control. The level of significance was set
at P=0.05.
DATA AVAILABILITY STATEMENT
The datasets generated for this study are available on request to
the corresponding author.
Frontiers in Endocrinology | www.frontiersin.org 7December 2019 | Volume 10 | Article 881
Bowman et al. Cardiac SNARE Proteins
ETHICS STATEMENT
The animal study was reviewed and approved by Glasgow
University Ethical Review Board.
AUTHOR CONTRIBUTIONS
PB designed and performed the experiments, analyzed the
data, prepared the figures, and wrote the first draft. GS
and GG conceived the study, obtained funding, designed the
study, and supervised the laboratory work. GS and GG edited
the manuscript.
FUNDING
This work was supported by a Ph.D. scholarship to PRTB
(FS/14/61/31284) and project grants (PG/18/47/33822)
from The British Heart Foundation to GG and
GS, and Diabetes UK (15/0005246 and 13/0004696
to GG).
ACKNOWLEDGMENTS
Data in this manuscript was submitted as part of PB’s Ph.D. thesis
to the University of Glasgow, 2019. We thank Dr. Anna White
and Dr. Augusto Montezano for supplying tissues and Dr. Jessica
Sadler for purified proteins.
SUPPLEMENTARY MATERIAL
The Supplementary Material for this article can be found
online at: https://www.frontiersin.org/articles/10.3389/fendo.
2019.00881/full#supplementary-material
REFERENCES
1. An D, Rodrigues B. Role of changes in cardiac metabolism in development of
diabetic cardiomyopathy. Am J Physiol Heart Circ Physiol. (2006) 291:1489–
506. doi: 10.1152/ajpheart.00278.2006
2. Resnick HE, Valsania P, Halter JB, Lin X. Relation of weight gain and weight
loss on subsequent diabetes risk in overweight adults. J Epidemiol Commun
Health. (2000) 54:596–602. doi: 10.1136/jech.54.8.596
3. Mazumder PK, O’Neill BT, Roberts MW, Buchanan J, Yun UJ, Cooksey
RC, et al. Impaired cardiac efficiency and increased fatty acid oxidation
in insulin-resistant Ob/Ob mouse hearts. Diabetes. (2004) 53:2366–74.
doi: 10.2337/diabetes.53.9.2366
4. Kannel WB, McGee DL. Diabetes and glucose tolerance as risk factors for
cardiovascular disease: the framingham study. Diabetes Care. (1979) 2:120–26.
doi: 10.2337/diacare.2.2.120
5. Boyer JK, Thanigaraj S, Schechtman KB, Pérez JE. Prevalence of ventricular
diastolic dysfunction in asymptomatic, normotensive patients with diabetes
mellitus. Am J Cardiol. (2004) 93:870–75. doi: 10.1016/j.amjcard.2003.12.026
6. Zahiti BF, Gorani DR, Gashi FB, Gjoka SB, Zahiti LB, Haxhiu BS, et al. Left
ventricular diastolic dysfunction in asymptomatic type 2 diabetic patients:
detection and evaluation by tissue doppler imaging. Acta Informat Med.
(2013) 21:120–23. doi: 10.5455/aim.2013.21.120-122
7. Poulsen MK, Henriksen JE, Dahl J, Johansen A, Gerke O, Vach W, et al.
Left ventricular diastolic function in type 2 diabetes mellitus prevalence and
association with myocardial and vascular disease. Circ Cardiovasc Imaging.
(2010) 3:24–31. doi: 10.1161/CIRCIMAGING.109.855510
8. Peterson LR, Herrero P, Schechtman KB, Racette SB, Waggoner AD, Kisrieva-
Ware Z, et al. Effect of obesity and insulin resistance on myocardial substrate
metabolism and efficiency in young women. Circulation. (2004) 109:2191–96.
doi: 10.1161/01.CIR.0000127959.28627.F8
9. Deng J, Huang J, Lu L, Hung L. Impairment of cardiac insulin signaling
and myocardial contractile performance in high-cholesterol/fructose-
fed rats. Am J Physiol Heart Circ Physiol. (2007) 293:978–87.
doi: 10.1152/ajpheart.01002.2006
10. Rijzewijk LJ, van der Meer RW, Lamb HJ, de Jong, H. W. A. M., Lubberink
M, Romijn JA, et al. Altered myocardial substrate metabolism and decreased
diastolic function in nonischemic human diabetic cardiomyopathy. J Am
College Cardiol. (2009) 54:1524–32. doi: 10.1016/j.jacc.2009.04.074
11. Belke DD, Larsen TS, Gibbs EM, Severson DL. Altered metabolism
causes cardiac dysfunction in perfused hearts from diabetic (Db/Db)
mice. Am J Physiol Endocrinol Metabol. (2000) 279:1104–13.
doi: 10.1152/ajpendo.2000.279.5.E1104
12. Semeniuk LM, Kryski AJ, Severson DL. Echocardiographic assessment
of cardiac function in diabetic Db/Db and transgenic Db/Db -HGLUT4
mice. Am J Physiol Heart Circ Physiol. (2002) 283:H976–82.
doi: 10.1152/ajpheart.00088.2002
13. Koek HL, Soedamah-Muthu SS, Kardaun, J. W. P. F., Gevers E, de Bruin
A, Reitsma JB, et al. Short- and long-term mortality after acute myocardial
infarction: comparison of patients with and without diabetes mellitus. Eur J
Epidemiol. (2007) 22:883–88. doi: 10.1007/s10654-007-9191-5
14. Malmberg K, Ryden L, Hamstent A, Herlitz J, Waldenstrom A, Wedel
H. Effects of insulin treatment on cause-specific one-year mortality and
morbidity in diabetic patients with acute myocardial infarction. Eur Heart J.
(1996) 17:1337–44. doi: 10.1093/oxfordjournals.eurheartj.a015067
15. Marfella R, Di Filippo C, Portoghese M, Ferraraccio F, Rizzo MR, Siniscalchi
M, et al. Tight glycemic control reduces heart inflammation and remodeling
during acute myocardial infarction in hyperglycemic patients. J Am College
Cardiol. (2009) 53:1425–36. doi: 10.1016/j.jacc.2009.01.041
16. Fu F, Zhao K, Li J, Xu J, Zhang Y, Liu C, et al. Direct evidence that myocardial
insulin resistance following myocardial ischemia contributes to post-ischemic
heart failure. Sci Rep. (2015) 5:17927. doi: 10.1038/srep17927
17. Abel ED, Kaulbach HC, Tian R, Hopkins JCA, Duffy J, Doetschman T,
et al. Cardiac hypertrophy with preserved contractile function after selective
deletion of GLUT4 from the heart. J Clin Invest. (1999) 104:1703–14.
doi: 10.1172/JCI7605
18. Fischer Y, Thomas J, Sevilla L, Muñoz P, Becker C, Holman G, et al.
Insulin-induced recruitment of glucose transporter 4 (GLUT4) and GLUT1
in isolated rat cardiac myocytes. Evidence of the Existence of different
intracellular GLUT4 vesicle populations. J Biol Chem. (1997) 272:7085–92.
doi: 10.1074/jbc.272.11.7085
19. Bryant NJ, Gould GW. SNARE proteins underpin insulin-regulated GLUT4
traffic. Traffic. (2011) 12:657–64. doi: 10.1111/j.1600-0854.2011.01163.x
20. Foster LJ, Yaworsky K, Trimble WS, Klip A. SNAP23 promotes insulin-
dependent glucose uptake in 3T3-L1 adipocytes: possible interaction
with cytoskeleton. Am J Physiol Cell Physiol. (1999) 276:C1108–14.
doi: 10.1152/ajpcell.1999.276.5.C1108
21. Wong PPC, Daneman N, Volchuk A, Lassam N, Wilson MC, Klip A,
et al. Tissue distribution of SNAP-23 and its subcellular localization
in 3T3-L1 cells. Biochem Biophys Res Commun. (1997) 230:64–68.
doi: 10.1006/bbrc.1996.5884
22. Cheatham B, Volchuk A, Kahn CR, Wang L, Rhodes CJ, Klip A.
Insulin-stimulated translocation of GLUT4 glucose transporters requires
SNARE-complex proteins. Proc Natl Acad Sci USA. (1996) 93:15169–73.
doi: 10.1073/pnas.93.26.15169
23. Volchuk A, Wang Q, Ewart HS, Liu Z, He L, Bennett MK, et al.
Syntaxin 4 in 3T3-L1 adipocytes: regulation by insulin and participation
in insulin-dependent glucose transport. Mol Biol Cell. (1996) 7:1075–82.
doi: 10.1091/mbc.7.7.1075
Frontiers in Endocrinology | www.frontiersin.org 8December 2019 | Volume 10 | Article 881
Bowman et al. Cardiac SNARE Proteins
24. Sumitani S, Ramlal T, Liu Z, Klip A. Expression of syntaxin 4 in rat skeletal
muscle and rat skeletal muscle cells in culture. Biochem Biophys Res Commun.
(1995) 213:462–68. doi: 10.1006/bbrc.1995.2154
25. Volchuk A, Mitsumoto Y, He L, Liu Z, Habermann E, Trimble W,
et al. Expression of Vesicle-Associated Membrane Protein 2 (VAMP-2)-
synaptobrevin II and cellubrevin in rat skeletal muscle and in a muscle cell
line. Biochem J. (1994) 304:139–45. doi: 10.1042/bj3040139
26. Kawanishi M, Tamori Y, Okazawa H, Araki S, Shinoda H, Kasuga M. Role
of SNAP23 in insulin-induced translocation of GLUT4 in 3T3-L1 adipocytes.
Mediation of complex formation between syntaxin4 and VAMP2. J Biol Chem.
(2000) 275:8240–7. doi: 10.1074/jbc.275.11.8240
27. Kawaguchi T, Tamori Y, Kanda H, Yoshikawa M, Tateya S, Nishino
N, et al. The T-SNAREs Syntaxin4 and SNAP23 but Not v-SNARE
VAMP2 are indispensable to tether GLUT4 vesicles at the plasma
membrane in adipocyte. Biochem Biophys Res Commun. (2010) 391:1336–41.
doi: 10.1016/j.bbrc.2009.12.045
28. Perera HKI, Clarke M, Morris NJ, Hong W, Chamberlain LH, Gould GW.
Syntaxin 6 regulates Glut4 trafficking in 3T3-L1 adipocytes. Mol Biol Cell.
(2003) 14:2946–58. doi: 10.1091/mbc.e02-11-0722
29. Proctor KM, Miller SCM, Bryant NJ, Gould GW. Syntaxin 16 controls
the intracellular sequestration of GLUT4 in 3T3-L1 adipocytes. Biochem
Biophys Res Commun. (2006) 347:433–38. doi: 10.1016/j.bbrc.2006.
06.135
30. Olson AL, Knight JB, Pessin JE. Syntaxin 4, VAMP2, and/or
VAMP3/Cellubrevin are functional target membrane and vesicle SNAP
receptors for insulin-stimulated GLUT4 translocation in adipocytes. Mol Cell
Biol. (1997) 17:2425–35. doi: 10.1128/MCB.17.5.2425
31. Spurlin BA, Park S, Nevins AK, Kim JK, Thurmond DC. Syntaxin
4 transgenic mice exhibit enhanced insulin-mediated glucose uptake in
skeletal muscle. Diabetes. (2004) 53:2223–31. doi: 10.2337/diabetes.53.
9.2223
32. Yang C, Coker KJ, Kim JK, Mora S, Thurmond DC, Davis AC, et al. Syntaxin 4
heterozygous knockout mice develop muscle insulin resistance. J Clin Invest.
(2001) 107:1311–18. doi: 10.1172/JCI12274
33. Jain SS, Snook LA, Glatz JFC, Luiken, J. J. F. P., Holloway GP, Thurmond DC,
et al. Munc18c provides stimulus-selective regulation of GLUT4 but not fatty
acid transporter trafficking in skeletal muscle. FEBS Lett. (2012) 586:2428–35.
doi: 10.1016/j.febslet.2012.05.061
34. Fukuda N, Emoto M, Nakamori Y, Taguchi A, Miyamoto S, Uraki S,
et al. DOC2B: a novel Syntaxin-4 binding protein mediating insulin-
regulated GLUT4 vesicle fusion in adipocytes. Diabetes. (2009) 58:377–84.
doi: 10.2337/db08-0303
35. Griffin ME, Marcucci MJ, Cline GW, Bell K, Barucci N, Lee D, et al. Free
fatty acid-induced insulin resistance is associated with activation of protein
kinase C theta and alterations in the insulin signaling cascade. Diabetes. (1999)
48:1270–74. doi: 10.2337/diabetes.48.6.1270
36. Kim JK, Fillmore JJ, Sunshine MJ, Albrecht B, Higashimori T, Kim D, et al.
PKC-theta knockout mice are protected from fat-induced insulin resistance. J
Clin Invest. (2004) 114:823–27. doi: 10.1172/JCI200422230
37. Yamauchi T, Kamon J, Waki H, Terauchi Y, Kubota N, Hara K, et al.
The fat-derived hormone adiponectin reverses insulin resistance associated
with both lipoatrophy and obesity. Nat Med. (2001) 7:941–6. doi: 10.1038/
90984
38. Plomgaard P, Bouzakri K, Krogh-Madsen R, Mittendorfer B, Zierath
JR, Pedersen BK. Tumor necrosis factor-alpha induces skeletal
muscle insulin resistance in healthy human subjects via inhibition
of Akt substrate 160 phosphorylation. Diabetes. (2005) 54:2939–45.
doi: 10.2337/diabetes.54.10.2939
39. Maier VH, Melvin DR, Lister CA, Chapman H, Gould GW, Murphy
GJ. V- and t-SNARE protein expression in models of insulin resistance:
normalization of glycemia by rosiglitazone treatment corrects overexpression
of cellubrevin, vesicle-associated membrane Protein-2, and Syntaxin 4 in
skeletal muscle of zucker diabetic fatty rats. Diabetes. (2000) 49:618–25.
doi: 10.2337/diabetes.49.4.618
40. Schlaepfer IR, Pulawa LK, Ferreira LD, James DE, Capell WH, Eckel
RH. Increased expression of the SNARE accessory protein Munc18c
in lipid-mediated insulin resistance. J Lipid Res. (2003) 44:1174–81.
doi: 10.1194/jlr.M300003-JLR200
41. Foley KP, Klip A. Dynamic GLUT4 sorting through a Syntaxin-6
compartment in muscle cells is derailed by insulin resistance-causing
ceramide. Biol Open. (2014) 3:314–25. doi: 10.1242/bio.20147898
42. Lancha A, López-Garrido S, Rodríguez A, Catalán V, Ramírez B, Valent,í
V, et al. Expression of syntaxin 8 in visceral adipose tissue is increased
in obese patients with type 2 diabetes and related to markers of
insulin resistance and inflammation. Arch Med Res. (2015) 46:47–53.
doi: 10.1016/j.arcmed.2014.12.003
43. Schwenk RW, Angin Y, Steinbusch LKM, Dirkx E, Hoebers N, Coumans
WA, et al. Overexpression of VAMP3, but Not VAMP2, protects GLUT4
translocation in an in vitro model of cardiac insulin resistance. J Biol Chem.
(2012) 287:37530–9. doi: 10.1074/jbc.M112.363630
44. Mohseni R, ArabSadeghabadi Z, Ziamajidi N, Abbasalipourkabir R,
RezaeiFarimani A. Oral Administration of resveratrol-loaded solid lipid
nanoparticle improves insulin resistance through targeting expression
of SNARE proteins in adipose and muscle tissue in rats with type
2 diabetes. Nanoscale Res Lett. (2019) 14:227. doi: 10.1186/s11671-019-
3042-7
45. Rezaei Farimani A, Saidijam M, Goodarzi MT, Azari RY, Asadi S, Zarei S, et al.
Effect of resveratrol supplementation on the SNARE proteins expression in
adipose tissue of stroptozotocin-nicotinamide induced type 2 diabetic rats.
Iran J Med Sci. (2015) 40:248–55.
46. Schwenk RW, Dirkx E, Coumans WA, Bonen A, Klip A, Glatz JFC,
et al. Requirement for distinct vesicle-associated membrane proteins in
insulin- and AMP-Activated Protein Kinase (AMPK)-Induced Translocation
of GLUT4 and CD36 in cultured cardiomyocytes. Diabetologia. (2010)
53:2209–19. doi: 10.1007/s00125-010-1832-7
47. Peters CG, Miller DF, Giovannucci DR. Identification, localization and
interaction of SNARE proteins in atrial cardiac myocytes. J Mol Cell Cardiol.
(2006) 40:361–74. doi: 10.1016/j.yjmcc.2005.12.007
48. Hickson GRX, Chamberlain LH, Maier VH, Gould GW. Quantification
of SNARE protein levels in 3T3-L1 adipocytes: implications for insulin-
stimulated glucose transport. Biochem Biophys Res Commun. (2000) 270:841–
5. doi: 10.1006/bbrc.2000.2525
49. Bowman PRT. Regulation of glucose transport in cardiomyocytes. Ph.D. thesis.
The University of Glasgow 2019. (2019). Available online at: http://theses.gla.
ac.uk/41002/
50. Kraegen EW, Sowden JA, Halstead MB, Clark PW, Rodnick KJ, Chisholm
DJ, et al. Glucose transporters and in vivo glucose uptake in skeletal and
cardiac muscle: fasting, insulin stimulation and immunoisolation studies of
GLUT1 and GLUT4. Biochem J. (1993) 295 (Pt 1):287–93. doi: 10.1042/
bj2950287
51. Kobayashi K, Forte TM, Taniguchi S, Ishida BY, Oka K, Chan L. The
Db/Db mouse, a model for diabetic dyslipidemia: molecular characterization
and effects of western diet feeding. Metabol Clin Exp. (2000) 49:22–31.
doi: 10.1016/S0026-0495(00)90588-2
52. Carroll R, Carley AN, Dyck JRB, Severson DL. Metabolic effects of insulin on
cardiomyocytes from control and diabetic Db/Db mouse hearts. Am J Physiol
Endocrinol Metabol. (2005) 288:E900–6. doi: 10.1152/ajpendo.00491.2004
53. Panagia M, Schneider JE, Brown B, Cole MA, Clarke K. Abnormal function
and glucose metabolism in the type-2 diabetic Db/Db mouse. Can J Physiol
Pharmacol. (2007) 85:289–94. doi: 10.1139/Y07-028
54. Broderick TL, Jankowski M, Wang D, Danalache BA, Parrott CR, Gutkowska
J. Downregulation in GATA4 and downstream structural and contractile
genes in the Db/Db mouse heart. ISRN Endocrinol. (2012) 2012:736860.
doi: 10.5402/2012/736860
55. Dupuis J, Langenberg C, Prokopenko I, Saxena R, Soranzo N, Jackson
AU, et al. New genetic loci implicated in fasting glucose homeostasis
and their impact on type 2 diabetes risk. Nat Genet. (2010) 42:105–16.
doi: 10.1038/ng.520
56. Burke SJ, Batdorf HM, Burk DH, Noland RC, Eder AE, Boulos MS, et al.
Db/Db mice exhibit features of human type 2 diabetes that are not present in
weight-matched C57BL/6J mice fed a western diet. J Diabet Res. (2017) 2017:
8503754. doi: 10.1155/2017/8503754
57. Ouwens DM, Boer C, Fodor M, de Galan P, Heine RJ, Maassen JA,
et al. Cardiac dysfunction induced by high-fat diet is associated with
altered myocardial insulin signalling in rats. Diabetologia. (2005) 48:1229–37.
doi: 10.1007/s00125-005-1755-x
Frontiers in Endocrinology | www.frontiersin.org 9December 2019 | Volume 10 | Article 881
Bowman et al. Cardiac SNARE Proteins
58. Almabrouk TAM, White AD, Ugusman AB, Skiba DS, Katwan OJ,
Alganga H, et al. High fat diet attenuates the anticontractile activity
of aortic PVAT via a mechanism involving AMPK and reduced
adiponectin secretion. Front Physiol. (2018) 9: 51. doi: 10.3389/fphys.2018.
00051
59. Planavila A, Fernández-Solà J, Villarroya F. Cardiokines as modulators of
stress-induced cardiac disorders. Adv Prot Chem Struct Biol. (2017) 108:227–
56. doi: 10.1016/bs.apcsb.2017.01.002
60. Sadler JBA, Bryant NJ, Gould GW. Characterization of VAMP isoforms in
3T3-L1 adipocytes: implications for GLUT4 trafficking. Mol Biol Cell. (2015)
26:530–36. doi: 10.1091/mbc.E14-09-1368
Conflict of Interest: The authors declare that the research was conducted in the
absence of any commercial or financial relationships that could be construed as a
potential conflict of interest.
Copyright © 2019 Bowman, Smith and Gould. This is an open-access article
distributed under the terms of the Creative Commons Attribution License (CC BY).
The use, distribution or reproduction in other forums is permitted, provided the
original author(s) and the copyright owner(s) are credited and that the original
publication in this journal is cited, in accordance with accepted academic practice.
No use, distribution or reproduction is permitted which does not comply with these
terms.
Frontiers in Endocrinology | www.frontiersin.org 10 December 2019 | Volume 10 | Article 881
... This process is catalyzed by NSF and SNAPs. This assembly will destabilize the lipid bilayers and permits the ion channel to be directed to the plasma membrane ( Figure 2C) (173). Another contributor to the ion channel trafficking is ankyrin. ...
Article
Full-text available
Heart failure (HF) is recognized as an epidemic in the contemporary world, impacting around 1%–2% of the adult population and affecting around 6 million Americans. HF remains a major cause of mortality, morbidity, and poor quality of life. Several therapies are used to treat HF and improve the survival of patients; however, despite these substantial improvements in treating HF, the incidence of HF is increasing rapidly, posing a significant burden to human health. The total cost of care for HF is USD 69.8 billion in 2023, warranting a better understanding of the mechanisms involved in HF. Among the most serious manifestations associated with HF is arrhythmia due to the electrophysiological changes within the cardiomyocyte. Among these electrophysiological changes, disruptions in sodium and potassium currents’ function and trafficking, as well as calcium handling, all of which impact arrhythmia in HF. The mechanisms responsible for the trafficking, anchoring, organization, and recycling of ion channels at the plasma membrane seem to be significant contributors to ion channels dysfunction in HF. Variants, microtubule alterations, or disturbances of anchoring proteins lead to ion channel trafficking defects and the alteration of the cardiomyocyte’s electrophysiology. Understanding the mechanisms of ion channels trafficking could provide new therapeutic approaches for the treatment of HF. This review provides an overview of the recent advances in ion channel trafficking in HF.
... One possible explanation for altered GSV interactions with the PM in insulin resistance is changes in the abundance of proteins comprising the SNARE complex and/or regulators of SNARE complex formation. VAMP2, VAMP3, and STX4 were elevated in skeletal muscle tissues from zucker diabetic fatty rats [61] while VAMP5 and SNAP29 were increased in insulin-resistant cardiac tissue [62]. Since fusogenic SNARE complexes require specific subunit stoichiometry, altered SNARE protein abundance could lead to 'dead-end' complexes that are unable to facilitate fusion. ...
Article
Full-text available
Insulin-stimulated glucose uptake into muscle and adipose tissue is vital for maintaining whole-body glucose homeostasis. Insulin promotes glucose uptake into these tissues by triggering a protein phosphorylation signalling cascade, which converges on multiple trafficking processes to deliver the glucose transporter GLUT4 to the cell surface. Impaired insulin-stimulated GLUT4 translocation in these tissues underlies insulin resistance, which is a major risk factor for type 2 diabetes and other metabolic diseases. Despite this, the precise changes in insulin signalling and GLUT4 trafficking underpinning insulin resistance remain unclear. In this review, we highlight insights from recent unbiased phosphoproteomics studies, which have enabled a comprehensive examination of insulin signalling and have transformed our perspective on how signalling changes may contribute to insulin resistance. We also discuss how GLUT4 trafficking is disrupted in insulin resistance, and underline sites where signalling changes could lead to these trafficking defects. Lastly, we address several major challenges currently faced by researchers in the field. As signalling and trafficking alterations can be examined at increasingly high resolution, integrative approaches examining the two in combination will provide immense opportunities for elucidating how they conspire to cause insulin resistance.
... Phosphorylation of AKT can accelerate the transfer of GLUT4 from the cell to the cell membrane, where it fuses with the cell membrane and increases glucose uptake [68,69]. In contrast, insulin-regulated GLUT4 trafficking is mediated by the formation of specific SNARE complexes [70,71]. Recent studies have found that disturbances in SNARE complex function can alter steroid hormone production and central nervous system behavior, as well as peripheral metabolism, providing potential insights into the relationship between metabolic and neuropsychiatric disorders in humans. ...
Article
Full-text available
Alzheimer's disease is the most common form of neurodegenerative disease, and increasing evidence shows that insulin signaling has crucial roles in AD initiation and progression. In this study, we explored the effect and underlying mechanism of SQW, a representative formula for tonifying the kidney and promoting yang, on improving the cognitive function in a streptozotocin-induced model of AD rats. We investigated memory impairment in the AD rats by using the Morris water test. HE and Nissl staining were employed to observe the histomorphological changes in the hippocampal. Expression levels of NeuN and proteins related to Tau and apoptosis were measured using immunohistochemistry and Western blotting, respectively. Additionally, we performed RNA sequencing, and the selected hub genes were then validated by qRT-PCR. Furthermore, the protein expression levels of PI3K/AKT pathway-related proteins were detected by Western blot. We found that SQW treatment significantly alleviated learning and memory impairment, pathological damage, and apoptosis in rats, as evidenced by an increased level of NeuN and Bcl-2, and decreased phosphorylation of Tau, Bax, and Caspase-3 protein expression. SQW treatment reversed the expression of insulin resistance-related genes (Nr4a1, Lpar1, Bdnf, Atf2, and Ppp2r2b) and reduced the inhibition of the PI3K/AKT pathway. Our results demonstrate that SQW could contribute to neuroprotection against learning and memory impairment in rats induced by STZ through activation of the PI3K/AKT pathway.
... 63 Additionally, a recent study that aimed to uncover the role of SNAREs in cardiac insulin resistance and Glut4 trafficking failed to detect differential expression of Stx4 between control, C57BL/KsJ-lepr db / lepr db , or high-fat diet fed mice. 80 Conditional Stx4 KO mice have also been used to investigate Stx4's requirement in bone matrix deposition. 81,82 Despite these proposed roles for STX4, patient 1 did not exhibit metabolic or bone mineralization defects, the former of which is another common feature of SRS. ...
Article
Full-text available
Requirements for vesicle fusion within the heart remain poorly understood, despite the multitude of processes that necessitate proper intracellular trafficking within cardiomyocytes. Here, we show that Syntaxin 4 (STX4), a target-Soluble N-ethylmaleimide sensitive factor attachment receptor (t-SNARE) protein, is required for normal vertebrate cardiac conduction and vesicular transport. Two patients were identified with damaging variants in STX4. A patient with a homozygous R240W missense variant displayed biventricular dilated cardiomyopathy, ectopy and runs of non-sustained ventricular tachycardia, sensorineural hearing loss, global developmental delay, and hypotonia, while a second patient displayed severe pleiotropic abnormalities and perinatal lethality. CRISPR/Cas9-generated stx4 mutant zebrafish exhibited defects reminiscent of these patients’ clinical presentations, including linearized hearts, bradycardia, otic vesicle dysgenesis, neuronal atrophy, and touch insensitivity by three days post fertilization. Imaging of Vamp2+ vesicles within stx4 mutant zebrafish hearts showed reduced docking to the cardiomyocyte sarcolemma. Optical mapping of the embryonic hearts coupled with pharmacological modulation of Ca²⁺ handling together support that zebrafish stx4 mutants have a reduction in L-type Ca²⁺ channel modulation. Transgenic overexpression of zebrafish Stx4R241W, analogous to the first patient’s STX4R240W variant, indicated that the variant is hypomorphic. Thus, these data show an in vivo requirement for SNAREs in regulating normal embryonic cardiac function and that variants in STX4 are associated with pleiotropic human disease, including cardiomyopathy.
... Transportation is achieved by the fusion of a donor compartment with an acceptor compartment. Each fusion step involves SNARE proteins (soluble N-ethylmaleimide-sensitive factor attachment protein receptors) carried by both the donor vesicle membrane (one vesicle-membrane SNARE) and the acceptor membrane (three target-membrane SNAREs), which destabilize the lipid bilayers and allow the channel to move to the acceptor compartment when interacting [10]. Ion channel trafficking in the cytoplasm is coordinated by various Rab GTPases, monomeric GTPases of the Ras superfamily. ...
Article
Full-text available
Both inherited and acquired cardiac arrhythmias are often associated with the abnormal functional expression of ion channels at the cellular level. The complex machinery that continuously traffics, anchors, organizes, and recycles ion channels at the plasma membrane of a cardiomyocyte appears to be a major source of channel dysfunction during cardiac arrhythmias. This has been well established with the discovery of mutations in the genes encoding several ion channels and ion channel partners during inherited cardiac arrhythmias. Fibrosis, altered myocyte contacts, and post-transcriptional protein changes are common factors that disorganize normal channel trafficking during acquired cardiac arrhythmias. Channel availability, described notably for hERG and KV1.5 channels, could be another potent arrhythmogenic mechanism. From this molecular knowledge on cardiac arrhythmias will emerge novel antiarrhythmic strategies.
... Interestingly, soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins are important for neurotransmitter release and other functions in neurons and glial cells [22]. SNARE proteins are generally small proteins of around 100-300 aminoacids and they mediate the fusion of biological membranes by localizing both at the vesicular membrane and on the target membrane (for further details please refer to [23][24][25][26][27]. They share a common domain, the SNARE motif, which is normally of 60 aa in length. ...
Article
Full-text available
Neurodegenerative diseases are pathologies of the central and peripheral nervous systems characterized by loss of brain functions and problems in movement which occur due to the slow and progressive degeneration of cellular elements. Several neurodegenerative diseases are known such as Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis and many studies on the molecular mechanisms underlying these pathologies have been conducted. Altered functions of some key proteins and the presence of intraneuronal aggregates have been identified as responsible for the development of the diseases. Interestingly, the formation of the SNARE complex has been discovered to be fundamental for vesicle fusion, vesicle recycling and neurotransmitter release. Indeed, inhibition of the formation of the SNARE complex, defects in the SNARE-dependent exocytosis and altered regulation of SNARE-mediated vesicle fusion have been associated with neurodegeneration. In this review, the biological aspects of neurodegenerative diseases and the role of SNARE proteins in relation to the onset of these pathologies are described.
... As summarized above, the role of v-and t-SNARE palmitoylation in modulating GLUT4 trafficking and glucose uptake has been so far only explored in fat cells. However, adipocytes [108] and HEK cells [110] provide information concerning the PATs associated with VAMP2 and SNAP23, which are similarly expressed in the heart [114,115]. In adipocytes, VAMP2 is palmitoylated by the Golgi-localized DHHC7 ( Figure 5) [108]. ...
Article
Full-text available
In the heart, inhibition of the insulin cascade following lipid overload is strongly associated with contractile dysfunction. The translocation of fatty acid transporter CD36 (SR-B2) from intracellular stores to the cell surface is a hallmark event in the lipid-overloaded heart, feeding forward to intracellular lipid accumulation. Yet, the molecular mechanisms by which intracellularly arrived lipids induce insulin resistance is ill-understood. Bioactive lipid metabolites (diacyl-glycerols, ceramides) are contributing factors but fail to correlate with the degree of cardiac insulin resistance in diabetic humans. This leaves room for other lipid-induced mechanisms involved in lipid-induced insulin resistance, including protein palmitoylation. Protein palmitoylation encompasses the reversible covalent attachment of palmitate moieties to cysteine residues and is governed by protein acyl-transferases and thioesterases. The function of palmitoylation is to provide proteins with proper spatiotemporal localization, thereby securing the correct unwinding of signaling pathways. In this review, we provide examples of palmitoylations of individual signaling proteins to discuss the emerging role of protein palmitoylation as a modulator of the insulin signaling cascade. Second, we speculate how protein hyper-palmitoylations (including that of CD36), as they occur during lipid oversupply, may lead to insulin resistance. Finally, we conclude that the protein palmitoylation machinery may offer novel targets to fight lipid-induced cardiomyopathy.
Article
Full-text available
This review article offers a detailed examination of metabolic adaptations in pressure overload hypertrophic hearts, a condition that plays a pivotal role in the progression of heart failure with preserved ejection fraction (HFpEF) to heart failure with reduced ejection fraction (HFrEF). The paper delves into the complex interplay between various metabolic pathways, including glucose metabolism, fatty acid metabolism, branched-chain amino acid metabolism, and ketone body metabolism. In-depth insights into the shifts in substrate utilization, the role of different transporter proteins, and the potential impact of hypoxia-induced injuries are discussed. Furthermore, potential therapeutic targets and strategies that could minimize myocardial injury and promote cardiac recovery in the context of pressure overload hypertrophy (POH) are examined. This work aims to contribute to a better understanding of metabolic adaptations in POH, highlighting the need for further research on potential therapeutic applications.
Article
Pancreatic β-cell dysfunction is an early hallmark of type 1 diabetes mellitus. Among the potentially critical factors that cause β-cell dysfunction are cytokine attack, glucotoxicity, induction of endoplasmic reticulum (ER) or mitochondria stress. However, the exact molecular mechanism underlying β-cell's inability to maintain glucose homeostasis under severe stresses is unknown. This study used proinflammatory cytokines, thapsigargin, and rotenone in the presence of high concentration glucose to mimicking the conditions experienced by dysfunctional β-cells in human pancreatic islets, and profiled the alterations to the islet proteome with TMT-based proteomics. The results were further verified with label-free quantitative proteomics. The differentially expressed proteins under stress conditions reveal that immune related pathways are mostly perturbed by cytokines, while the respiratory electron transport chains and protein processing in ER pathways by rotenone. Thapsigargin together with high glucose induces dramatic increases of proteins in lipid synthesis and peroxisomal protein import pathways, with energy metabolism and vesicle secretion related pathways downregulated. High concentration glucose, on the other hand, alleviated complex I inhibition induced by rotenone. Our results contribute to a more comprehensive understanding of the molecular events involved in β-cell dysfunction.
Article
Full-text available
Abstract In the current study, we developed resveratrol (RES)-loaded solid lipid nanoparticle (SLN-RES) in order to improve insulin resistance through the upregulation of SNARE protein complex in rats with type 2 diabetes. The SLN-RES characteristics include the following: the average size of 248 nm, the zeta potential of − 16.5 mV, and 79.9% RES entrapment efficiency. The release profile of SLN-RES showed an initial burst followed by a sustained release in natural condition. Infrared spectroscopy results revealed good incorporation of RES into core SLN. Spherical nanoparticle with less aggregation was observed under electronic microscopic examination. Oral administration of SLN-RES prevented weight loss and showed better hypoglycemic effect than RES. Serum oxidative stress status was restored to the normal level by SLN-RES. Furthermore, expression of synaptosomal-associated protein 23 (Snap23), syntaxin-4 (Stx4), and vesicle-associated membrane protein 2 (Vamp2) as the major elements of SNARE protein complex were reduced by SLN-RES more significantly than RES treatment in muscle tissue. However, SLN-RES has a similar effect to RES treatment in adipose tissue. Taken together, our results revealed SLN-RES could be a modern and interestingly therapeutic approach for the improvement of insulin resistance through targeting the expression of Snap23, Stx4, and Vamp2 in adipose and muscle tissues.
Article
Full-text available
Background and aim: Perivascular adipose tissue (PVAT) positively regulates vascular function through production of factors such as adiponectin but this effect is attenuated in obesity. The enzyme AMP-activated protein kinase (AMPK) is present in PVAT and is implicated in mediating the vascular effects of adiponectin. In this study, we investigated the effect of an obesogenic high fat diet (HFD) on aortic PVAT and whether any changes involved AMPK. Methods: Wild type Sv129 (WT) and AMPKα1 knockout (KO) mice aged 8 weeks were fed normal diet (ND) or HFD (42% kcal fat) for 12 weeks. Adiponectin production by PVAT was assessed by ELISA and AMPK expression studied using immunoblotting. Macrophages in PVAT were identified using immunohistochemistry and markers of M1 and M2 macrophage subtypes evaluated using real time-qPCR. Vascular responses were measured in endothelium-denuded aortic rings with or without attached PVAT. Carotid wire injury was performed and PVAT inflammation studied 7 days later. Key results: Aortic PVAT from KO and WT mice was morphologically indistinct but KO PVAT had more infiltrating macrophages. HFD caused an increased infiltration of macrophages in WT mice with increased expression of the M1 macrophage markers Nos2 and Il1b and the M2 marker Chil3. In WT mice, HFD reduced the anticontractile effect of PVAT as well as reducing adiponectin secretion and AMPK phosphorylation. PVAT from KO mice on ND had significantly reduced adiponectin secretion and no anticontractile effect and feeding HFD did not alter this. Wire injury induced macrophage infiltration of PVAT but did not cause further infiltration in KO mice. Conclusions: High-fat diet causes an inflammatory infiltrate, reduced AMPK phosphorylation and attenuates the anticontractile effect of murine aortic PVAT. Mice lacking AMPKα1 phenocopy many of the changes in wild-type aortic PVAT after HFD, suggesting that AMPK may protect the vessel against deleterious changes in response to HFD.
Article
Full-text available
To understand features of human obesity and type 2 diabetes mellitus (T2D) that can be recapitulated in the mouse, we compared C57BL/6J mice fed a Western-style diet (WD) to weight-matched genetically obese leptin receptor-deficient mice ( db/db ). All mice were monitored for changes in body composition, glycemia, and total body mass. To objectively compare diet-induced and genetic models of obesity, tissue analyses were conducted using mice with similar body mass. We found that adipose tissue inflammation was present in both models of obesity. In addition, distinct alterations in metabolic flexibility were evident between WD-fed mice and db/db mice. Circulating insulin levels are elevated in each model of obesity, while glucagon was increased only in the db/db mice. Although both WD-fed and db/db mice exhibited adaptive increases in islet size, the db/db mice also displayed augmented islet expression of the dedifferentiation marker Aldh1a3 and reduced nuclear presence of the transcription factor Nkx6.1. Based on the collective results put forth herein, we conclude that db/db mice capture key features of human T2D that do not occur in WD-fed C57BL/6J mice of comparable body mass.
Article
Full-text available
A close link between heart failure (HF) and systemic insulin resistance has been well documented, whereas myocardial insulin resistance and its association with HF are inadequately investigated. This study aims to determine the role of myocardial insulin resistance in ischemic HF and its underlying mechanisms. Male Sprague-Dawley rats subjected to myocardial infarction (MI) developed progressive left ventricular dilation with dysfunction and HF at 4 wk post-MI. Of note, myocardial insulin sensitivity was decreased as early as 1 wk after MI, which was accompanied by increased production of myocardial TNF-α. Overexpression of TNF-α in heart mimicked impaired insulin signaling and cardiac dysfunction leading to HF observed after MI. Treatment of rats with a specific TNF-α inhibitor improved myocardial insulin signaling post-MI. Insulin treatment given immediately following MI suppressed myocardial TNF-α production and improved cardiac insulin sensitivity and opposed cardiac dysfunction/remodeling. Moreover, tamoxifen-induced cardiomyocyte-specific insulin receptor knockout mice exhibited aggravated post-ischemic ventricular remodeling and dysfunction compared with controls. In conclusion, MI induces myocardial insulin resistance (without systemic insulin resistance) mediated partly by ischemia-induced myocardial TNF-α overproduction and promotes the development of HF. Our findings underscore the direct and essential role of myocardial insulin signaling in protection against post-ischemic HF.
Article
Full-text available
Unlabelled: Using isolated rat cardiomyocytes we have examined: 1) the effect of insulin on the cellular distribution of glucose transporter 4 (GLUT4) and GLUT1, 2) the total amount of these transporters, and 3) the co-localization of GLUT4, GLUT1, and secretory carrier membrane proteins (SCAMPs) in intracellular membranes. Insulin induced 5.7- and 2.7-fold increases in GLUT4 and GLUT1 at the cell surface, respectively, as determined by the nonpermeant photoaffinity label [3H]2-N-[4(1-azi-2,2,2-trifluoroethyl)benzoyl]-1, 3-bis-(D-mannos-4-yloxy)propyl-2-amine. The total amount of GLUT1, as determined by quantitative Western blot analysis of cell homogenates, was found to represent a substantial fraction ( approximately 30%) of the total glucose transporter content. Intracellular GLUT4-containing vesicles were immunoisolated from low density microsomes by using monoclonal anti-GLUT4 (1F8) or anti-SCAMP antibodies (3F8) coupled to either agarose or acrylamide. With these different immunoisolation conditions two GLUT4 membrane pools were found in nonstimulated cells: one pool with a high proportion of GLUT4 and a low content in GLUT1 and SCAMP 39 (pool 1) and a second GLUT4 pool with a high content of GLUT1 and SCAMP 39 (pool 2). The existence of pool 1 was confirmed by immunotitration of intracellular GLUT4 membranes with 1F8-acrylamide. Acute insulin treatment caused the depletion of GLUT4 in both pools and of GLUT1 and SCAMP 39 in pool 2. In conclusion: 1) GLUT4 is the major glucose transporter to be recruited to the surface of cardiomyocytes in response to insulin; 2) these cells express a high level of GLUT1; and 3) intracellular GLUT4-containing vesicles consist of at least two populations, which is compatible with recently proposed models of GLUT4 trafficking in adipocytes.
Article
Full-text available
Both syntaxin4 and VAMP2 are implicated in insulin regulation of glucose transporter-4 (GLUT4) trafficking in adipocytes as target (t) soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNARE) and vesicle (v)-SNARE proteins, respectively, which mediate fusion of GLUT4-containing vesicles with the plasma membrane. Synaptosome-associated 23-kDa protein (SNAP23) is a widely expressed isoform of SNAP25, the principal t-SNARE of neuronal cells, and colocalizes with syntaxin4 in the plasma membrane of 3T3-L1 adipocytes. In the present study, two SNAP23 mutants, SNAP23-ΔC8 (amino acids 1 to 202) and SNAP23-ΔC49 (amino acids 1 to 161), were generated to determine whether SNAP23 is required for insulin-induced translocation of GLUT4 to the plasma membrane in 3T3-L1 adipocytes. Wild-type SNAP23 (SNAP23-WT) promoted the interaction between syntaxin4 and VAMP2 both in vitro andin vivo. Although SNAP23-ΔC49 bound to neither syntaxin4 nor VAMP2, the SNAP23-ΔC8 mutant bound to syntaxin4 but not to VAMP2. In addition, although SNAP23-ΔC8 bound to syntaxin4, it did not mediate the interaction between syntaxin4 and VAMP2. Moreover, overexpression of SNAP23-ΔC8 in 3T3-L1 adipocytes by adenovirus-mediated gene transfer inhibited insulin-induced translocation of GLUT4 but not that of GLUT1. In contrast, overexpression of neither SNAP23-WT nor SNAP23-ΔC49 in 3T3-L1 adipocytes affected the translocation of GLUT4 or GLUT1. Together, these results demonstrate that SNAP23 contributes to insulin-dependent trafficking of GLUT4 to the plasma membrane in 3T3-L1 adipocytes by mediating the interaction between t-SNARE (syntaxin4) and v-SNARE (VAMP2).
Article
Full-text available
Glucose uptake by muscles and fat cells is carried out by the GLUT4 system. Isoforms of the SNAP23, syntaxin-4 and VAMP-2 play an important role in regulating GLUT-4 trafficking and fusion in adipocytes. The changes of SNARE proteins levels and thus impaired GLUT-4 displacement can be one of the etiological causes of type 2 diabetes. Due to changes in the expression of these proteins in diabetes, the aim of this study was to investigate the effect of the natural compound resveratrol with anti-diabetic properties on impaired expression of SNARE proteins in type 2 diabetes. Forty male Wistar rats were used in this study. Type 2 diabetes was induced by administering a single dose of streptozotocin and nicotinamide. The expression of SNAP-23, syntaxin-4 and VAMP-2 proteins were assessed using real-time qRT-PCR. Also, some biochemical parameters were examined, including fasting blood glucose, insulin levels and insulin resistance. The results of this study showed that, resveratrol supplementation increased blood insulin level, reduced the fasting blood glucose, and improved the insulin resistance. In addition, resveratrol supplementation increased the expression of SNAP-23, syntaxin-4 and VAMP-2 proteins that involved in GLUT-4 transport in adipose tissue of diabetic rats. Final results showed that SNARE proteins expression is significantly reduced in diabetic rats and treatment with resveratrol supplementation is associated with the increased expression of these proteins.
Chapter
Almost 30 years ago, the protein, atrial natriuretic peptide, was identified as a heart-secreted hormone that provides a peripheral signal from the myocardium that communicates to the rest of the organism to modify blood pressure and volume under conditions of heart failure. Since then, additional peripheral factors secreted by the heart, termed cardiokines, have been identified and shown to coordinate this interorgan cross talk. In addition to this interorgan communication, cardiokines also act in an autocrine/paracrine manner to play a role in intercellular communication within the myocardium. This review focuses on the roles of newly emerging cardiokines that are mainly increased in stress-induced cardiac diseases. The potential of these cardiokines as clinical biomarkers for diagnosis and prognosis of cardiac disorders is also discussed.