ArticlePDF Available

γ-Aminbuturic Acid A Receptor Mitigates Homocysteine-Induced Endothelial Cell Permeability

Authors:

Abstract and Figures

Many cerebrovascular disorders are accompanied by an increased homocysteine (Hcy) levels. We have previously shown that acute hyperhomocysteinemia (HHcy) leads to an increased microvascular permeability in the mouse brain. Hcy competitively binds to gamma -aminbuturic acid (GABA) receptors and may increase vascular permeability by acting as an excitatory neurotransmitter. However, the role of GABA-A (GABA(A)) receptor in Hcy-induced endothelial cell (EC) permeability remains unclear. In the present study we attempted to determine the role of GABA(A) receptor and the possible mechanisms involved in Hcy-induced EC layer permeability. Mouse aortic and brain ECs were grown in Transwells and treated with 50 mu M Hcy in the presence or absence of GABA(A)-specific agonist muscimol. Role of matrix metalloproteinase-9 (MMP-9) was determined using its activity inhibitor GM-6001. Involvement of extracellular signal-regulated kinase (ERK) signaling was assessed using its kinase activity inhibitors PD98059 or U0126. EC permeability to the known content of bovine serum albumin (BSA)-conjugated with Alexa Flour-488 was assessed by measuring fluorescence intensity of the solutes in the Transwell's lower chambers. It was found that Hcy induced the formation of filamentous actin (F-actin). Hcy-induced EC permeability to BSA was significantly decreased by GABA and muscimol treatments. Presence of MMP-9 or ERK kinase activity inhibitors restored the Hcy-induced EC permeability to its baseline level. The mediation BSA leakage through the ECs was further confirmed in the experiments where Hcy-induced alterations in transendothelial electrical resistance of confluent ECs were assessed. The data suggest that Hcy increases EC layer permeability through inhibition of GABA(A) receptor and F-actin formation, in part, by transducing ERK and MMP-9 activation.
Content may be subject to copyright.
γ-Aminbuturic Acid A Receptor Mitigates Homocysteine-Induced
Endothelial Cell Permeability
Neetu Tyagi, Karni S. Moshal, Suresh C. Tyagi, and David Lominadze
Department of Physiology and Biophysics, University of Louisville, Louisville, Kentucky, USA
Abstract
Many cerebrovascular disorders are accompanied by an increased homocysteine (Hcy) levels. We
have previously shown that acute hyperhomocysteinemia (HHcy) leads to an increased microvascular
permeability in the mouse brain. Hcy competitively binds to γ -aminbuturic acid (GABA) receptors
and may increase vascular permeability by acting as an excitatory neurotransmitter. However, the
role of GABA-A (GABAA) receptor in Hcy-induced endothelial cell (EC) permeability remains
unclear. In the present study we attempted to determine the role of GABAA receptor and the possible
mechanisms involved in Hcy-induced EC layer permeability. Mouse aortic and brain ECs were grown
in Transwells and treated with 50 μM Hcy in the presence or absence of GABAA-specific agonist
muscimol. Role of matrix metalloproteinase-9 (MMP-9) was determined using its activity inhibitor
GM-6001. Involvement of extracellular signal-regulated kinase (ERK) signaling was assessed using
its kinase activity inhibitors PD98059 or U0126. EC permeability to the known content of bovine
serum albumin (BSA)-conjugated with Alexa Flour-488 was assessed by measuring fluorescence
intensity of the solutes in the Transwell's lower chambers. It was found that Hcy induced the formation
of filamentous actin (F-actin). Hcy-induced EC permeability to BSA was significantly decreased by
GABA and muscimol treatments. Presence of MMP-9 or ERK kinase activity inhibitors restored the
Hcy-induced EC permeability to its baseline level. The mediation BSA leakage through the ECs was
further confirmed in the experiments where Hcy-induced alterations in transendothelial electrical
resistance of confluent ECs were assessed. The data suggest that Hcy increases EC layer permeability
through inhibition of GABAA receptor and F-actin formation, in part, by transducing ERK and
MMP-9 activation.
Keywords
Albumin Leakage; Endothelial Gap Formation; ERK; F-Actin; Muscimol
Homocysteine (Hcy) is a homologue of the naturally occurring amino acid, cysteine. Normal
concentration of Hcy in plasma is in the range of 5 to 10 μM. Increased blood level of Hcy is
known as hyperhomocysteinemia (HHcy). There are three ranges of HHcy: moderate (10 to
30 μM), intermediate (31 to 100 μM), and severe (>100 μM). Increased blood levels of Hcy
Copyright © Informa Healthcare USA, Inc.
Address correspondence to David Lominadze, PhD, Department of Physiology and Biophysics, Health Sciences Center, A-1115,
University of Louisville, Louisville, KY 40292, USA. dglomi01@louisville.edu.
Publisher's Disclaimer: Full terms and conditions of use: http://www.informaworld.com/terms-and-conditions-of-access.pdf This
article maybe used for research, teaching and private study purposes. Any substantial or systematic reproduction, re-distribution, re-
selling, loan or sub-licensing, systematic supply or distribution in any form to anyone is expressly forbidden. The publisher does not give
any warranty express or implied or make any representation that the contents will be complete or accurate or up to date. The accuracy
of any instructions, formulae and drug doses should be independently verified with primary sources. The publisher shall not be liable for
any loss, actions, claims, proceedings, demand or costs or damages whatsoever or howsoever caused arising directly or indirectly in
connection with or arising out of the use of this material.
NIH Public Access
Author Manuscript
Endothelium. Author manuscript; available in PMC 2010 February 10.
Published in final edited form as:
Endothelium. 2007 ; 14(6): 315. doi:10.1080/10623320701746164.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
have been linked to increased risk of premature coronary artery disease, stroke, and
thromboembolism (Lentz 2005; Lee et al. 2006). Elevation of blood Hcy content coincides
with an increased blood concentration of well-accepted inflammatory markers such as C-
reactive protein (Hackam and Anand 2003), interleukin-6 (Araki et al. 2005), and fibrinogen
(Zoccali et al. 2002). Because of this association, it was postulated that an increase in Hcy
content may promote vascular inflammation (Lentz 2005). In addition, it was shown that
increased level of Hcy may cause interleukin-6 accumulation in the monocytes (van Aken et
al. 2000), suggesting an association of HHcy and an enhanced formation of interleukin-6, the
latter being involved in synthesis of another inflammatory agent fibrinogen (Vasse et al.
1996). However, according to the recent Glasgow Myocardial Infarction Study, correlation of
Hcy with the inflammatory markers such as fibrinogen, C-reactive protein, and interleukin-6
was not significant (Woodward et al. 2006), suggesting that, within the population range,
increasing Hcy contents have no effect on the systemic inflammatory response. Therefore, it
can be stated that Hcy is an independent risk factor for cardiovascular diseases and stroke
(Seshadri et al. 2002; Tyagi 1999).
Many cardiovascular diseases, e.g., hypertension and diabetes, are considered inflammatory
diseases. Increased microvascular permeability, also typical to these diseases, is one of the
manifestations of inflammation. A net loss of blood plasma components into the interstitium
causes edema. Albumin, the most abundant plasma protein, helps maintain the oncotic pressure
and protects endothelial barrier integrity by its interaction with glycocalyx (Huxley and Curry
1985, 1987). Thus, changes in albumin transport through the endothelial cell (EC) layer may
cause significant damage to tissue and induce inflammatory responses (Mehta and Malik
2006).
It has been shown that HHcy induced by folate depletion caused increased arterial permeability
and stiffness in rats (Symons et al. 2002, 2006). The results of our previous study suggest that
an acute increase in blood Hcy content enhances leakage of pial microvessels in mice
(Lominadze et al. 2006). Although the role of Hcy-induced matrix metalloproteinase-9
(MMP-9) activation in vascular remodeling is well known, the mechanism of Hcy-induced
increased microvascular permeability was not clear. Along with extracellular matrix (ECM)
degradation, tissue plasminogen activator causes proteolytic shedding of γ-aminobutyric acid
(GABA) receptors (Mataga et al. 2002; Frey et al. 1996). GABA receptors play a significant
role in brain microvascular permeability, which if inhibited, alters the neuronal environment
and leads to ECM degradation and edema (Lee et al. 1995; Limmroth et al. 1996; Fruscella et
al. 2001; Lazzarini et al. 2001). Hcy competes with GABA receptors and acts as an excitatory
neurotransmitter. Therefore, an increase in blood Hcy level may attenuate the function of
GABA receptor, leading to the disruption of EC layer integrity. In the present study, we tested
the hypothesis that high levels of Hcy may increase EC leakage to albumin through extracellular
signal regulated kinase (ERK) signaling and filamentous actin (F-actin) formation. The role of
GABA-A (GABAA) receptor in Hcy-induced EC permeability is shown.
METHODS
Materials and Reagents
Bovine serum albumin (BSA) 1-palmitoyl-sn-glycero-3-phosphocholine (LPC), Hcy,
fibronectin, and muscimol (GABAA receptor–specific agonist) were purchased from Sigma
(St. Louis, MO). Specific inhibitors of the mitogen-activated protein kinase known as
extracellular signal-regulated kinase (ERK), kinase, PD98059 (2-amino-3-methoxyflavone),
and U0126 (1,4-diamino-2,3-dicyano-1,4-bis[2-aminophenylthio]butadiene), and GM-6001
(MMP activity blocker) were purchased from Calbiochem (La Jolla, CA). Monoclonal
antibody to phosphorylated ERK1/2 (p44/42) was purchased from Cell Signaling Technology
(Beverly, MA) and the antibody to ERK2 was purchased from Santa Cruz Biotechnology
Tyagi et al. Page 2
Endothelium. Author manuscript; available in PMC 2010 February 10.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
(Santa Cruz, CA). Horseradish peroxidase (HRP)-conjugated antibody was obtained from
Cappel Laboratories (Durham, NC). BSA conjugated with Alexa Flour-488 dye (BSA-488),
Alexa Flour-594 Phalloidin (300 U), and a secondary antibody conjugated with Texas Red dye
were purchased from Molecular Probes (Eugene, OR).
Endothelial Cell Culture
Mouse aortic endothelial cells (MAECs) were a generous gift from Kathleen Bove, Stratton
VA Medical Center, Albany, NY. The endothelial nature of the cells was verified by uptake
of acylated low-density lipoprotein and positive staining for CD-31 (Lincoln et al. 2003).
Mouse brain endothelial cells (MBECs) were purchased from American Type Culture
Collection (ATCC, Manassas, VA) at 21th passage. The endothelial nature of these cells was
confirmed by the observed expression of von Willebrand factor and uptake of fluorescently
labeled low-density lipoprotein (Montesano et al. 1990). The MAECs were grown in
Dulbecco's modified Eagle's medium/Ham's F-12 50/50 mix (DMEM/F12–50/50; Cellgrow,
Mediatech, Herndon, VA), whereas MBEC were grown in DMEM (ATCC). Both culture
media were supplemented with 10% fetal bovine serum (HyClone, Logan, UT) 1% L-glutamine
and 1% penicillin-streptomycin (Gibco, Grand Island, NY). Cells were grown at 37°C at 5%
CO2 in a humidified environment. For the experimentation, MAECs and MBECs were used
at the 10th and 22nd passages, respectively.
Endothelial Cell Permeability Assays
Albumin leakage through endothelial cell monolayer was assessed according to the method
described earlier (Tyagi et al. 2007b). Transwell permeable supports (Corning, Corning, NJ)
with polycarbonate membranes (Nuclepore Track-Etch; 6.5 mm in diameter, 0.4-μm pore size,
and pore density of 108/cm2) were coated with fibronectin for 1 h. The membranes were seeded
with MAECs or MBECs and the cells were grown to confluence. To confirm the cell confluence
and the presence of an intact monolayer on the membranes, cells in a separate test well (not
membrane) were monitored by a light microscope. Cells in the test membranes were labeled
with fluorescence (2 ,7-bis(2-carboxyethyl)-5-(and-6)-carboxyfluorescein, acetoxymethyl
ester [BCECF, AM]; Molecular Probes, Eugene, OR) and observed under a microscope (Carl
Zeiss Axiovert-100, objective 10×) with a fluorescence filter (488-nm excitation and 516-nm
emission) for absence of visible gaps in the cell monolayer. The permeability assays were done
after confirming that the cells in the test wells and Transwell membranes were fully confluent
and formed an intact monolayer.
Unlabeled BSA was added to each well to maintain its concentration similar to a normal plasma
concentration of albumin (440 μM) and maintain its activity coefficient close to that in blood
(Rivas et al. 1999). The surface levels of solutions in the luminal (200 μL) and abluminal (600
μL) compartments of the Transwells were the same. The experimental set-up is similar to that
used by Cooper et al. (1987), but the presence of albumin-mediated osmotic pressure in the
present study makes it closer to in vivo conditions (Tyagi et al. 2007b).
For the permeability assay, cells were washed with phosphate-buffered saline (PBS) and
incubated with various doses of Hcy (5, 25, or 50 μM) for 24 h, or with 50 μM of Hcy for 1,
12, or 24 h. To determine a possible role of GABAA receptor, MMPs, and the mitogen-activated
protein kinase kinase (MEK) signaling in Hcy-induced EC permeability, the following
substances were added to the Transwells: Hcy (50 μM) with muscimol (50 μM), Hcy (50 μM)
with MEK inhibitors (PD98059 or U0126; 50 μM each), Hcy (50 μM) with GABA (50 μM),
or Hcy (50 μM) with GM6001 (1 μM).
To determine the effect of GABA, muscimol, GM6001, and MEK inhibitors on EC
permeability to albumin, the following substances were added to other Transwells without Hcy:
Tyagi et al. Page 3
Endothelium. Author manuscript; available in PMC 2010 February 10.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
GABA (50 μM), muscimol (50 μM), GM6001 (1 μM), PD98059 (50 μM), or U0126 (50 μM).
Cells incubated with medium alone were used as a control group. BSA-488 (3 mg/mL) was
added to each of the wells described above and experiments were performed five times in
duplicate for each treatment.
The cells were incubated in humidified conditions at 37°C. After incubation, media samples
were collected from lower and upper wells. Fluorescence intensity of the samples was measured
by a microplate reader (SpectraMax M2; Molecular Devices, Sunnyvale, CA) with excitation
at 494 nm and emission at 518 nm. Results are expressed as fluorescence intensity units (FIU)
and presented as a percent of the control values.
In additional experiments, permeability of the Nuclepore membranes alone to albumin was
compared to albumin leakage through the EC layer in Tranwells. The results showed that
albumin leakage through the membrane alone (583 ± 34 FIU) was about nine times greater
than that (66 ± 12 FIU) in the presence of the cells.
Transendothelial electrical resistance measurement was preformed using an electrical cell-
substrate impedance system (ECIS; Applied Biophysics, Troy, NY). ECs were seeded onto
evaporated gold microplates of the ECIS. The cells were grown to a confluent monolayer that
covered the microelectrodes of the system's 8-well chamber (Tiruppathi et al.1992). Effect of
Hcy (50 μM) was determined in the presence or absence of muscimol (50 μM), MEK inhibitors
(PD98059 or U0126; 50 μM each), GABA (50 μM), or with GM6001 (1 μM). To determine
the effect of GABA, muscimol, GM6001, and MEK inhibitors on transendothelial electrical
resistance of the cells, the following substances were added to wells without Hcy: GABA (50
μM), muscimol (50 μM), GM6001 (1 μM), PD98059 (50 μM), or U0126 (50 μM). Cells
incubated with medium alone were used as a control group. Resistance values from each
microelectrode were collected and plotted as relative resistance versus time.
F-Actin Formation Assay
Formation of F-actin in cultured ECs was studied according to the method described earlier
(Lominadze et al. 2004, 2006; Tyagi et al. 2007). Briefly, MAECs and MBECs were grown
until confluent in 8-well coverglass plates coated with fibronectin. The cells were washed and
incubated with or without PD98059 or U0126 (50 μM each), muscimol (50 μM), or GABA
(50 μM), followed by Hcy (50 μM) treatment for 24 h. To determine the effect of GABA,
muscimol, PD98059, or U0126 on F-actin formation, the separate groups of cells were
incubated with following substances without Hcy: GABA (50 μM), muscimol (50 μM),
PD98059 (50 μM), or U0126 (50 μM). Cells incubated with medium alone were used as a
control. After the treatments the cells were washed twice with PBS and incubated with Alexa
Flour-594 Phalloidin (10 U) and LPC (100 μg/mL, dissolved in 3.7% formaldehyde) for 30
min at 4°C in the dark (Lominadze et al. 2006). After incubation, the cells were washed with
PBS and digital images of intracellular F-actin were taken with the confocal microscope
(Olympus FV1000, objective 100×) using a HeNe-G laser (596 nm) to excite the dye, whereas
emission was observed above 620 nm. The images were compared for the formation of
individual stress fibers and presence of actin foci (Lominadze et al. 2006).
In separate experiments, Hcy-induced formation of F-actin in ECs were compared to F-actin
formation induced by thrombin. The cells were treated with or without 50 μMHcy for 24 h and
0.5 U/mL thrombin for 15 min (Qiao et al. 1995;Trepat et al. 2005; Tyagi et al. 2007) and
analyzed for intracellular F-actin formation using confocal microscopy.
Hcy-induced formation of F-actin fibers was assessed for each well by analyzing the total
fluorescence intensity in four random fields with image analysis software (Image-Pro Plus;
Media Cybernetics). Four experiments were done to study Hcy-induced F-actin formation and
Tyagi et al. Page 4
Endothelium. Author manuscript; available in PMC 2010 February 10.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
three experiments were done to study Hcy- and thrombin-induced F-actin formation. All the
experiments were done in duplicate (two wells per experimental group). Results are expressed
as a percent of the control values.
Western Blot Analysis for ERK Phosphorylation
Hcy-induced phosphorylation of ERK was studied using a method described previously
(Moshal et al. 2006). Briefly, MAECs were grown in 6-well plates (Corning, Corning, NY)
until they become 80% to 90% confluent. Before experimentation, cells were serum starved
by depriving fetal bovine serum to 0.01% in the medium for 16 h. Then cells were incubated
with one of the following: Hcy (5 or 50 μM), Hcy (50 μM) with muscimol (50 μM), Hcy (50
μM) with MEK inhibitors (PD98059 or U0126; 50 μM each), or Hcy (50 μM) with GABA (50
μM) at 37°C for 24 h. Other cells were incubated with one of the following: muscimol (50
μM), PD98059 or U0126 (50 μM each), or GABA (50 μM) at 37°C for 24 h. Cells incubated
with low-serum medium were used as a control.
After incubation, the cells were washed twice with ice-cold 1× PBS and lysed with ice-cold
lysis buffer (composition: Tris·HCl [50 mM], NaCl [150 mM], Triton X-100 [1%], and EGTA
[1 mM], pH 7.4) supplemented with phenylmethylsulfonyl fluoride (1 mM), leupeptin (1 μg/
mL), sodium orthovan-date (200 μM), and aprotinin (1 μg/mL). Cell lysates were assayed for
protein concentration using Bradford assay (Bradford 1976). Equal amount of protein was
resolved on 10% sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE)
and blotted onto a polyvinylidene difluoride (PVDF) membrane. After being transferred, blots
were washed with Tris-buffered saline with 0.1% Tween-20 (TBST) for 5 min at room
temperature and blocked with 5% BSA in TBST for 1 h at room temperature. The blots were
then incubated with polyclonal anti-rabbit phospho-ERK1/2 antibody (1:1000 dilution)
overnight at 4°C with gentle agitation. The blots were washed three times (8-min wash each
time) with TBST and incubated with appropriate secondary antibody (1:3000 dilution).
Proteins of interest were immuno detected using an enhanced chemiluminescence (ECL) plus
kit (Amersham Biosciences, Piscataway, NJ). The membranes were stripped and reprobed for
ERK2 as loading control. The blots were analyzed with Gel-Pro Analyzer software (Media
Cybernetics, Silver Spring, MD) as described earlier (Lominadze et al. 2002). The protein
expression intensity was assessed by integrated optical density (IOD), i.e., the area of the band
in the lane profile. To account for possible differences in the protein load, the results of the
measurements are presented as a ratio of IOD of each band under the study (phosphorylated
ERK1/2) to the IOD of the respective total protein (total ERK2).
Statistics
All data are expressed as mean ± SEM. The experimental groups were compared by one-way
analysis of variance (ANOVA). If ANOVA indicated a significant difference (p < .05), Tukey's
multiple comparison test was used to compare group means. Differences were considered
statistically signifi-cant if p < .05.
RESULTS
Hcy induced a dose-dependant increase in the permeability of the EC layer to albumin as
determined by the fluorescence intensity of albumin that leaked though MAECs into the lower
wells of the Transwells (Figure 1). The permeability of the MAECs to albumin, induced by
the highest dose of Hcy (50 μM), was increased (128% ± 9%, 186% ± 25%, 303% ± 34 % of
control) with treatment time (1, 12, and 24 h, respectively). The permeability of MAEC layer
to albumin induced by Hcy (50 μM) during 24 h was decreased significantly by GABA,
muscimol, GM6001, and MEK inhibitors (PD98059 or U0126) (Figure 1). In the absence of
Tyagi et al. Page 5
Endothelium. Author manuscript; available in PMC 2010 February 10.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Hcy, treatment of MAECs with GABA, muscimol, GM-6001, or MEK inhibitors (PD98059
or U0126) had no effect on albumin leakage through MAEC layer (Table 1).
The enhanced permeability of the MBECs to albumin, induced by Hcy (50 μM), was decreased
significantly by GABA, muscimol, MEK inhibitors (PD98059 or U0126), and GM6001
(Figure 2). In the absence of Hcy, treatment of MBECs with GABA, muscimol, MEK inhibitors
(PD98059 or U0126), or GM-6001 had no effect on MBEC albumin leakage (Table 2).
In separate experiments, Hcy treatment significantly decreased transendothelial electrical
resistance compared to untreated control MBECs (Figure 3). This decrease in resistance was
significantly less in the presence of muscimol and GABA (Figure 3). Treatment of the MBECs
with GABA and muscimol had no effect on transendothelial electrical resistance (Figure 3).
Hcy caused a dose-dependant increase of F-actin formation in MAECs (Figure 4). The
formation of F-actin, induced by the highest dose of Hcy (50 μM), was significantly decreased
by GABA, muscimol, or by MEK inhibitors (PD98059 or U0126) (Figure 4). Formation of F-
actin in the MAECs treated with GABA, muscimol, or with MEK inhibitors (PD98059 or
U0126) alone was not different from that in control group (Table 3). The treatment of ECs with
Hcy (50 μM) for 24 h caused the formation of F-actin (160% ± 6% of control), which was
similar to that (141% ± 4% of control) induced by thrombin (0.5 U/mL for 15 min) (Figure
4A). In the parallel series of experiments, treatment of MBECs with Hcy (50 μM) for 24 h
caused the formation of F-actin, which was not different from that induced by thrombin (0.5
U/ml for 15 min) (Figure 5).
Hcy induced a dose-dependant increase of ERK phosphorylation in MAECs that was
significantly decreased in the presence of muscimol (Figure 6). Treatment of MAECs with
muscimol alone did not alter ERK phosphorylation in the ECs compared to that in the control
group (Figure 6).
DISCUSSION
The present study shows that pathological concentrations of Hcy (<12 μM) can enhance
albumin leakage through an EC monolayer. These results coincide with our data showing that
Hcy increases albumin leakage of pial vessels in mice (Lominadze et al. 2006). It is known
that thrombin-induced intercellular gaps are accompanied by formation of F-actin (Qiao et al.
1995). In the present study, the treatment of the ECs with higher than normal dose of Hcy (50
μM) for 24 h caused F-actin formation to the level that was caused by thrombin (0.5 U/mL 15
min; Figure 4), which is known to increase F-actin formation (Trepat et al. 2005). These data
suggest that an increased concentration of Hcy in the blood stream has a role in increasing EC
permeability similar to thrombin.
Increased content of Hcy may enhance albumin leakage by inducing the formation of F-actin
(Figures 4 and 5), which may cause stiffening of the cells and opening of interendothelial
junctions (IEJs). Role of Hcy on mouse brain IEJs was clearly shown using ECIS (Figure 3),
where, the effect of Hcy was significantly diminished by GABA or muscimol. The results of
the present study also coincide with our previous results, where we showed that Hcy may affect
EC by activating MMPs (Lominadze et al. 2006; Moshal et al. 2006). The present results show
a restoration of Hcy-induced albumin leakage through EC layer in the presence of MMP
inhibitor (GM6001), which confirms the role of MMPs in Hcy-induced microvascular leakage
(Lominadze et al. 2006). Changes in MMPs may cause subendothelial matrix remodeling by
altering collagen/elastin ratio (Shastry et al. 2006) and target focal adhesion molecules and cell
junction molecules at the basal side of the ECs, such as cadherins and/or connexins (Mehta
and Malik 2006).
Tyagi et al. Page 6
Endothelium. Author manuscript; available in PMC 2010 February 10.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Significant decrease in Hcy-induced albumin leakage and F-actin formation by GABA or its
agonist, muscimol, suggests that Hcy may directly affect EC properties through binding to
GABAA receptor. This binding suppressed the activity of GABAA receptor (Shastry et al.
2006), making Hcy an excitatory neurotransmitter. Our studies show that mouse EC also
express GABAA receptor, and its activity can be suppressed by Hcy (Tyagi et al. 2007a). The
results of the present study suggest that the activation of GABAA receptor may prevent Hcy-
induced EC leakage and therefore highlight the functional role of GABAA receptor in
endothelium and its possible involvement in Hcy-induced EC layer permeability.
Mitogen-activated protein kinase kinase (MEK) is a protein kinase that is phosphorylated and
activated by Raf. Once activated, MEK phosphorylates and activates ERK (Kolch 2000).
PD9805 and U0126 used in the present study are chemically unrelated MEK/ERK inhibitors
with different mechanisms of action (Alessi et al. 1995; Favata et al. 1998). Whereas PD98059
binds to the inactive forms of MEK and prevents its activation by upstream activators such as
Raf (Alessi et al. 1995), U0126 binds to the activated form of MEK (Favata et al. 1998). It is
known that formation of F-actin can occur through ERK signaling (Bourguignon et al. 2005;
Chandrasekar et al. 2003). Furthermore, ERK signaling pathway is involved in expression of
GABAA receptor (Bulleit and Hsieh 2000; Kalluri and Ticku 2002). Our data show that Hcy-
induced albumin leakage may be regulated by ERK signaling (Figure 6). Furthermore, these
results, in combination with the data showing decreased albumin leakage through EC layer in
the presence of GABA or muscimol, suggest that competitive binding of Hcy to GABAA
receptor on EC surface involves the activation of MEK/ERK signal. In our previous study, we
showed that increased levels of Hcy enhance phosphorylation of ERK and activate MMP-9
(Moshal et al. 2006). The present study indicates the role of MMPs in Hcy-induced albumin
leakage through the EC layer. Increased content of Hcy enhances EC layer permeability, which
involves inhibition of GABAA receptor activity and formation of F-actin through ERK
signaling. Our data related to the muscimol-induced increase ERK phosphorylation confirm
the result by others showing that muscimol activates ERK signaling (Obrietan et al. 2002).
However, muscimol-induced ERK phosphorylation was not as robust as the one induced by
50 μM Hcy. The treatment of the cells with muscimol prior to addition of Hcy (50 μM) did not
increase ERK phosphorylation to the extent that was caused by the same dose of Hcy. This
suggests that msucimol competes with Hcy for GABAA receptor binding sites and induces
ERK phosphorylation.
Increased blood Hcy concentration may increase binding of Hcy to endothelial GABAA
receptors. Cytoskeletal ezrinradixin-moesin (ERM) family protein radixin has been identified
as an essential clustering factor that anchors GABAA receptor with actin cytoskeleton of a cell
(Tsukita and Yonemura 1999; Loebrich et al. 2006). A role of radixin in ERK signaling pathway
has been suggested (Jung et al. 2005). In the present study the role of radixin was not evaluated;
however, the role of GABAA receptor in radixin-mediated ERK signaling pathway can't be
ruled out during Hcy-induced endothelial monolayer leakage, which may cause the formation
of F-actin in ECs. F-actin formation may increase the rigidity of the cells, widen the IEJ, and
may even increase gaps between the cells (Ehringer et al. 1999; Gordon et al. 2005; Lominadze
et al. 2004, 2006). The enlarged IEJ gaps may be enough to allow albumin (diameter ~5 nm)
to pass though the EC monolayer.
Increased blood Hcy concentration is typical for many cardiovascular and cerebrovascular
diseases (Seshadri et al. 2002; Tyagi et al. 1999). Almost all of these diseases are accompanied
with increased ECs permeability. We provide the evidence that elevated levels of Hcy have a
role in increasing EC layer permeability through its binding to endothelial GABAA receptor
and suppressing its activity.
Tyagi et al. Page 7
Endothelium. Author manuscript; available in PMC 2010 February 10.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Limitations of the Present Study
The results of the present study show that elevated content of Hcy increases EC layer
permeability to albumin through IEJs. However, the design of our experiments did not permit
us to rule out transcellular migration of albumin (Mehta and Malik 2006). In addition to
affecting IEJs, Hcy may alter transcytosis affecting caveolae-involved an absorptive or fluid-
phase pathways, or formation of transendothelia channels. The experiments to address the role
of Hcy in transendothelial albumin transport are in progress.
Acknowledgments
This work was supported in part by the American Heart Association (AHA), National, SDG (grant 0235317N), NIH
(grant HL-080394 to DL, grants HL-71010 and NS-051568 to SCT), and the AHA, postdoctoral training grant (award
0625579B) to KSM.
REFERENCES
Alessi D, Cuenda A, Cohen P, Dudley D, Saltiel A. PD 098059 is a specific inhibitor of the activation
of mitogen-activated protein kinase kinase in vitro and in vivo. Journal of Biological Chemistryn
1995;270:27489–27494.
Araki A, Hosoi T, Orimo H, Ito H. Association of plasma homocysteine with serum interleukin-6 and C-
peptide levels in patients with type 2 diabetes. Metabolism 2005;54:809–814. [PubMed: 15931619]
Bourguignon L, Gilad E, Rothman K, Peyrollier K. Hyaluronan-CD44 interaction with IQGAP1
promotes Cdc42 and ERK signaling, leading to actin binding, Elk-1/estrogen receptor transcriptional
activation, and ovarian cancer progression. Journal of Biological Chemistry 2005;280:11961–11972.
[PubMed: 15655247]
Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein
utilizing the principle of protein-dye binding. Analytical Biochemistry 1976;72:248–254. [PubMed:
942051]
Bulleit RF, Hsieh T. MEK inhibitors block BDNF-dependent and -independent expression of GABA(A)
receptor subunit mRNAs in cultured mouse cerebellar granule neurons. Brain Research and
Developmental Brain Research 2000;119:1–10.
Chandrasekar N, Mohanam S, Lakka SS, Dinh DH, Olivero WC, Gujrati M, Rao JS. Glial cell-induced
endothelial morphogenesis is inhibited by interfering with extracellular signal-regulated kinase
signaling. Clinical Cancer Research 2003;9:2342–2349. [PubMed: 12796404]
Cooper J, Del Vecchio P, Minnear F, Burhop K, Selig W, Garcia J, Malik A. Measurement of albumin
permeability across endothelial monolayers in vitro. Journal of Applied Physiology 1987;62:1076–
1083. [PubMed: 3571065]
Ehringer W, Yamany S, Steier K, Farag A, Roisen F, Dozier A, Miller F. Quantitative image analysis of
F-actin in endothelial cells. Microcirculation 1999;6:291–303. [PubMed: 10654280]
Favata M, Horiuchi K, Manos E, Daulerio A, Stradley D, Feeser W, Van Dyk D, Pitts W, Earl R, Hobbs
F, Copeland R, Magolda R, Scherle P, Trzaskos J. Identification of a novel inhibitor of mitogen-
activated protein kinase kinase. Journal of Biological Chemistry 1998;273:18623–18632. [PubMed:
9660836]
Frey U, Muller M, Kuhl D. A different form of long-lasting potentiation revealed in tissue plasminogen
activator mutant mice. Journal of Neuroscience 1996;16:2057–2063. [PubMed: 8604050]
Fruscella P, Sottocorno M, Di BM, Diomede L, Piccardi N, Cagnotto A, Grossi G, Romano M, Mennini
T, Roma G. 1,5-Benzodiazepine tricyclic derivatives exerting anti-inflammatory effects in mice by
inhibiting interleukin-6 and prostaglandinE(2)production. Pharmacological Research 2001;43:445–
452. [PubMed: 11394936]
Gordon S, Lominadze D, Saari J, Lentsch A, Schuschke D. Impaired deformability of copper-deficient
neutrophils. Experimental Biology and Medicine 2005;230:543–548. [PubMed: 16118404]
Hackam DG, Anand SS. Emerging risk factors for atherosclerotic vascular disease: a critical review of
the evidence. Journal of the American Medical Association 2003;290:932–940. [PubMed: 12928471]
Tyagi et al. Page 8
Endothelium. Author manuscript; available in PMC 2010 February 10.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Huxley V, Curry F. Albumin modulation of capillary permeability: test of an adsorption mechanism.
American Journal of Physiology 1985;248:H264–H273. [PubMed: 3871592]
Huxley V, Curry F. Effect of superfusate albumin on single capillary hydraulic conductivity. American
Journal of Physiology 1987;252:H395–H401. [PubMed: 3492926]
Jung JR, Kim H, Jeun SS, Lee JY, Koh EJ, Ji C. The phosphorylation status of merlin is important for
regulating the Ras-ERK pathway. Molecules and Cells 2005;20:196–200. [PubMed: 16267393]
Kalluri HS, Ticku MK. Role of GABA(A) receptors in the ethanol-mediated inhibition of extracellular
signal-regulated kinase. European Journal of Pharmacology 2002;451:51–54. [PubMed: 12223228]
Kolch W. Meaningful relationships: The regulation of the Ras/Raf/MEK/ERK pathway by protein
interactions. Biochemical Journal 2000;351(Pt 2):289–305. [PubMed: 11023813]
Lazzarini R, Malucelli BE, Palermo-Neto J. Reduction of acute inflammation in rats by diazepam: role
of peripheral benzodiazepine receptors and corticosterone. Immunopharmacology and
Immunotoxicology 2001;23:253–265. [PubMed: 11417852]
Lee KW, Hill JS, Walley KR, Frohlich JJ. Relative value of multiple plasma biomarkers as risk factors
for coronary artery disease and death in an angiography cohort. Canadian Medical Association
Journal 2006;174:461–466. [PubMed: 16477056]
Lee WS, Limmroth V, Ayata C, Cutrer FM, Waeber C, Yu X, Moskowitz MA. Peripheral GABAA
receptor-mediated effects of sodium valproate on dural plasma protein extravasation to substance P
and trigeminal stimulation. British Journal of Pharmacology 1995;116:1661–1667. [PubMed:
8564234]
Lentz SR. Mechanisms of homocysteine-induced atherothrombosis. Journal of Thrombosis and
Haemostasis 2005;3:1646–1654. [PubMed: 16102030]
Limmroth V, Lee WS, Moskowitz MA. GABAA-receptor-mediated effects of progesterone, its ring-A-
reduced metabolites and synthetic neuroactive steroids on neurogenic oedema in the rat meninges.
British Journal of Pharmacology 1996;117:99–104. [PubMed: 8825349]
Lincoln DW 2nd, Larsen AM, Phillips PG, Bove K. Isolation of murine aortic endothelial cells in culture
and the effects of sex steroids on their growth. In Vitro Cell and Developmental Biology of Animals
2003;39:140–145.
Loebrich S, Bahring R, Katsuno T, Tsukita S, Kneussel M. Activated radixin is essential for GABAA
receptor alpha5 subunit anchoring at the actin cytoskeleton. EMBO Journal 2006;25:987–999.
[PubMed: 16467845]
Lominadze D, Roberts A, Tyagi N, Moshal KS, Tyagi S. Homocysteine causes cerebrovascular leakage
in mice. American Journal of Physiology Heart and Circulatory Physiology 2006;290:H1206–H1213.
[PubMed: 16258031]
Lominadze D, Saari J, Percival S, Schuschke D. Proinflammatory effects of copper deficiency on
neutrophils and lung endothelial cells. Immunology and Cell Biology 2004;82:231–238. [PubMed:
15186252]
Lominadze D, Schuschke D, Joshua I, Dean W. Increased ability of erythrocytes to aggregate in
spontaneously hypertensive rats. Clinical and Experimental Hypertension 2002;24:397–406.
[PubMed: 12109779]
Mataga N, Nagai N, Hensch TK. Permissive proteolytic activity for visual cortical plasticity. Proceedings
of the National Academy of Sciences of the United States of America 2002;99:7717–7721. [PubMed:
12032349]
Mehta D, Malik A. Signaling mechanisms regulating endothelial permeability. Physiological Reviews
2006;86:279–367. [PubMed: 16371600]
Montesano R, Pepper MS, Mohle-Steinlein U, Risau W, Wagner EF, Orci L. Increased proteolytic activity
is responsible for the aberrant morphogenetic behavior of endothelial cells expressing the middle T
oncogene. Cell 1990;62:435–445. [PubMed: 2379237]
Moshal KS, Sen U, Tyagi N, Henderson B, Steed M, Ovechkin AV, Tyagi SC. Regulation of
homocysteine-induced MMP-9 by ERK1/2 pathway. American Journal of Physiology and Cellular
Physiology 2006;290:C883–C891.
Obrietan K, Gao XB, Van Den Pol AN. Excitatory actions of GABA increase BDNF expression via a
MAPK-CREB-dependent mechanism—a positive feedback circuit in developing neurons. Journal
of Neurophysiology 2002;88:1005–1015. [PubMed: 12163549]
Tyagi et al. Page 9
Endothelium. Author manuscript; available in PMC 2010 February 10.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Qiao R, Yan W, Lum H, Malik A. Arg-Gly-Asp peptide increases endothelial hydraulic conductivity:
comparison with thrombin response. American Journal of Physiology Cellular Physiology
1995;269:C110–C117.
Rivas G, Fernandez J, Minton A. Direct observation of the self-association of dilute proteins in the
presence of inert macromolecules at high concentration via tracer sedimentation equilibrium: Theory,
experiment, and biological significance. Biochemistry 1999;38:9379–9388. [PubMed: 10413513]
Seshadri S, Beiser A, Selhub J, Jacques PF, Rosenberg IH, D'Agostino RB, Wilson PW, Wolf PA. Plasma
homocysteine as a risk factor for dementia and Alzheimer's disease. New England Journal of
Medicine 2002;346:476–483. [PubMed: 11844848]
Shastry S, Tyagi N, Moshal K, Lominadze D, Hayden M, Tyagi S. GABA receptors ameliorate Hcy-
mediated integrin shedding and constrictive collagen remodeling in microvascular endothelial cells.
Cellular Biochemistry and Biophysics 2006;45:157–165.
Symons JD, Mullick AE, Ensunsa JL, Ma AA, Rutledge JC. Hyperhomocysteinemia evoked by folate
depletion: effects on coronary and carotid arterial function. Arteriosclerosis, Thrombosis, and
Vascular Biology 2002;22:772–780.
Symons JD, Zaid UB, Athanassious CN, Mullick AE, Lentz SR, Rutledge JC. Influence of folate on
arterial permeability and stiffness in the absence or presence of hyperhomocysteinemia.
Arteriosclerosis, Thrombosis, and Vascular Biology 2006;26:814–818.
Tiruppathi C, Malik AB, Del Vecchio PJ, Keese CR, Giaever I. Electrical method for detection on
endothelial cell shape change in real time: Assessment of endothelial barrier function. Proceedings
of the National Academy of Sciences of the United States of America 1992;89:7919–7923. [PubMed:
1518814]
Trepat X, Grabulosa M, Buscemi L, Rico F, Farre R, Navajas D. Thrombin and histamine induce
stiffening of alveolar epithelial cells. Journal of Applied Physiology 2005;98:1567–1574. [PubMed:
15557012]
Tsukita S, Yonemura S. Cortical actin organization: lessons from ERM (ezrin/radixin/moesin) proteins.
Journal of Biological Chemistry 1999;274:34507–34510. [PubMed: 10574907]
Tyagi N, Lominadze D, Gillespie W, Moshal KS, Rosenberger DS, Steed M, Sen U, Tyagi SC.
Differential expression of the ?-aminobutyric acid receptor A (GABAA) and its subunits in various
tissue. Clinical Chemistry and Laboratory Medicine. 2007a (in press).
Tyagi N, Roberts AM, Dean WL, Tyagi SC, Lominadze D. Fibrinogen induces endothelial cell
permeability. Molecular and Cellular Biochemistry. 2007b (In press).
Tyagi SC. Homocyst(e)ine and heart disease: Pathophysiology of extra-cellular matrix. Clinical and
Experimental Hypertension 1999;21:181–198. [PubMed: 10225475]
van Aken BE, Jansen J, van Deventer SJ, Reitsma PH. Elevated levels of homocysteine increase IL-6
production in monocytic Mono Mac 6 cells. Blood Coagulation and Fibrinolysis 2000;11:159–164.
[PubMed: 10759009]
Vasse M, Paysant J, Soria J, Collet J, Vannier J, Soria C. Regulation of fibrinogen biosynthesis by
cytokines, consequences on the vascular risk. Haemostasis 1996;26:331–339. [PubMed: 8979138]
Woodward M, Rumley A, Rumley A, Rumley C, Lewington S, Morrison CE, Lowe GD. The association
between homocysteine and myocardial infarction is independent of age, sex, blood pressure,
cholesterol, smoking and markers of inflammation: the Glasgow Myocardial Infarction Study. Blood
Coagulation and Fibrinolysis 2006;17:1–5. [PubMed: 16607071]
Zoccali C, Mallamaci F, Maas R, Benedetto FA, Tripepi G, Malatino LS, Cataliotti A, Bellanuova I,
Boger R. Left ventricular hypertrophy, cardiac remodeling and asymmetric dimethylarginine
(ADMA) in hemodialysis patients. Kidney International 2002;62:339–345. [PubMed: 12081596]
Tyagi et al. Page 10
Endothelium. Author manuscript; available in PMC 2010 February 10.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
FIG. 1.
Hcy-induced albumin leakage through the mouse aortic endothelial cell monolayer.
Fluorescence intensity of bovine serum albumin conjugated with Alexa Flour-488 detected in
lower chambers of Transwells. *p < .05 versus 5 μM of Hcy. †p < .05 versus the lower dose
of Hcy. #p < .05 versus 50 μMof Hcy. Number of experiments n = 5 for all groups.
Tyagi et al. Page 11
Endothelium. Author manuscript; available in PMC 2010 February 10.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
FIG. 2.
Hcy-induced albumin leakage through the mouse brain microvascular endothelial cell
monolayer. Fluorescence intensity of bovine serum albumin conjugated with Alexa Flour-488
detected in lower chambers of Transwells. Hcy, GABA, muscimol, and MEK inhibitors,
PD98059 and U0126, were used at concentration of 50 μM each, whereas GM6001 was 1
μM. * p < .05 versus Hcy. Number of experiments n = 4 for all groups.
Tyagi et al. Page 12
Endothelium. Author manuscript; available in PMC 2010 February 10.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
FIG. 3.
Changes in relative resistance of the confluent mouse brain endothelial cell layers after
treatment with Hcy (•), GABA and Hcy (), muscimol and Hcy (), GABA (), and muscimol
(). Cells in the control group (*) were grown in medium alone. Hcy, GABA, and muscimol
were used at concentration of 50 μM each. Number of experiments n = 4 for all groups.
Tyagi et al. Page 13
Endothelium. Author manuscript; available in PMC 2010 February 10.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
FIG. 4.
Hcy-induced F-actin formation in mouse aortic endothelial cells. (A) Examples of images of
Hcy- and thrombin-induced F-actin formation (objective 100×). (B) Total fluorescence
intensity changes of F-actin staining induced by Hcy (5 and 50 μM) and inhibition of 50 μM
Hcy effect by GABA, muscimol, PD98059, and U0126 (50 μM each). * p < .05 versus 5 μM
of Hcy. # p < .05 versus 50 μM of Hcy. Each data point represents the results of duplicate
determinations from four separate experiments (n = 4).
Tyagi et al. Page 14
Endothelium. Author manuscript; available in PMC 2010 February 10.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
FIG. 5.
Hcy-induced F-actin formation in mouse brain endothelial cells. (A) Examples of images of
Hcy- and thrombin-induced F-actin formation (objective 100×). (B)Total fluorescence
intensity changes of F-actin staining induced by 50 μM of Hcy (24 h) and 0.5 U/ml of thrombin
(15 min). Each data point represents the results of duplicate determinations from four separate
experiments (n = 4).
Tyagi et al. Page 15
Endothelium. Author manuscript; available in PMC 2010 February 10.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
FIG. 6.
Hcy-induced ERK phosphorylation in MAECs. (A) A representative Western Blot for Hcy-
induced ERK1/2 phosphorylation and its inhibition in the presence of muscimol is shown
(top). Membranes were reprobed for total ERK2 (bottom). Phosphorylation of ERK in the cells
treated with medium alone was used as a control group. (B) ERK1/2 phosphorylation induced
by 50 μMHcy. The data are presented as ratios of integrated optical density (IOD) of each
phoshorylated ERK1/2 band to IOD of the respective total ERK2. Bars represent averages ±
SE from three different experiments (n = 3). * p < .05 versus control; #p < .05 versus 50 μM
of Hcy.
Tyagi et al. Page 16
Endothelium. Author manuscript; available in PMC 2010 February 10.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Tyagi et al. Page 17
TABLE 1
Effect of GABA, muscimol, GM6001, PD98059, or U0126 on albumin leakage through the mouse aortic
endothelial cell monolayer
GABA (50 μM) Muscimol (50 μM) GM6001 (1 μM) PD98059 (50 μM) U0126 (50 μM)
115 ± 13 121 ± 19 111 ± 12 108 ± 8 124 ± 21
Note. Data are presented as percent of control. Number of experiments n = 4.
Endothelium. Author manuscript; available in PMC 2010 February 10.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Tyagi et al. Page 18
TABLE 2
Effect of GABA, muscimol, PD98059, U0126, or GM6001 on albumin leakage through the mouse brain
endothelial cell monolayer
GABA (50 μM) Muscimol (50 μM) PD98059 (50 μM) U0126 (50 μM) GM6001 (1 μM)
107 ± 12 101 ± 11 108 ± 8 111 ± 13 121 ± 14
Note. Data are presented as percent of control. Number of experiments n = 4.
Endothelium. Author manuscript; available in PMC 2010 February 10.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Tyagi et al. Page 19
TABLE 3
Effect of GABA, muscimol, PD98059, U0126, or GM6001 on F-actin formation in the mouse aortic endothelial
cells
(50 μM) GABA (50 μM) Muscimol (50 μM) PD98059 (50 μM) U0126
109 ± 6 111 ± 6 98 ± 1 94 ± 3
Note. Data are presented as percent of control. Number of experiments n = 4.
Endothelium. Author manuscript; available in PMC 2010 February 10.
... Chromosomal DNA was isolated and the 16S rRNA gene was amplified by PCR [3]. The strains were identified by DNA sequencing (Sango Biotech, China) according to the method of Tyagi, N. et al. [16]. ...
Article
Full-text available
Probiotics can effectively modulate host immune responses and prevent gastrointestinal diseases. The objective of this study was to investigate the probiotic characteristics of Lactobacillus brevis KU15152 isolated from kimchi and its protective potential against intestinal inflammation induced by Staphylococcus aureus lipoteichoic acid (aLTA). L. brevis KU15152 exhibited a high survival rate in artificial gastric and bile environments. Additionally, the adhesion capability of the strain to HT-29 cells was higher than that of L. rhamnosus GG. L. brevis KU15152 did not produce harmful enzymes, such as β-glucuronidase, indicating that it could be used as a potential probiotic. The anti-inflammatory potential of L. brevis KU15152 was determined in HT-29 cells. Treatment with L. brevis KU15152 suppressed the production of interleukin-8 without inducing significant cytotoxicity. The downregulatory effects of L. brevis KU15152 were involved in the suppression of nuclear factor-kappa B activation mediated by the extracellular signal-regulated kinase and Akt signaling pathways. Collectively, these data suggest that L. brevis KU15152 can be used in developing therapeutic and prophylactic products to manage and treat aLTA-induced intestinal damage.
... Chromosomal DNA was isolated and the 16S rRNA gene was amplified by PCR [3]. The strains were identified by DNA sequencing (Sango Biotech, China) according to the method of Tyagi, N. et al. [16]. ...
Article
Full-text available
Lactobacillus reuteri NK33 (NK33) and Bifidobacterium adolescentis NK98 (NK98) alleviate immobilization stress-induced depression. To understand gut microbiota-mediated mechanism of NK33 and NK98 against depression, we examined their effects on Escherichia coli K1 (K1)-induced depression and gut dysbiosis in mice. NK33, NK98, and their mixtures (1:1, 4:1, and 9:1) mitigated K1-induced depression and colitis. NK33 and NK98 additively or synergistically increased BDNF+/NeuN+ cell populations and suppressed NF-κB action in the hippocampus. They alleviated gut dysbiosis by reducing the Proteobacteria population and increasing the Clostridia population. These results suggest that NK33 and NK98 may alleviate depression and colitis by ameliorating gut dysbiosis.
... Knockout studies in mouse exhibited that PDCD2 is indispensable for viability and self-renewal of embryonic stem cells (Mu et al., 2010). Furthermore, mouse PDCD2 protein does not bind to DNA as a transcriptional factor, but rather, physically interacts with host cell factor 1 that controls numerous phases of the cell cycle (Scarr and Sharp, 2002;Tyagi et al., 2007). However, in most of the animals, the regulatory role of Zfrp8/PDCD2 in cell proliferation remains to be elucidated. ...
Article
Programmed cell death 2 (PDCD2) is a highly conserved eukaryotic protein indispensable for various physiological processes such as cell proliferation, development, and apoptosis. In the present study, we identified a Zinc finger protein RP-8 from the silkworm, Bombyx mori (BmZfrp8), the ortholog of PDCD2 protein. The quantitative real-time PCR analysis revealed the ubiquitous distribution of BmZfrp8 in the different tissues; however, the gene's transcription level was highest in those of the silk gland, testis, and ovary. Additionally, the expression levels of BmZfrp8 were unequal on different days of embryonic development, and it reached the highest level on the 5th day of early development. The challenge with pathogens influenced the expression level of BmZfrp8 in both hemocyte and fat body when compared with the control. Administration of 20-hydroxyecdysone significantly enhanced the BmZfrp8 expression in hemocyte. The knock-down of BmZfrp8 by double-stranded RNA suppressed the expression of developmental pathway associated genes as well as cell cycle-associated genes. Furthermore, the RNAi treated cells also showed cell cycle arrest compared to the control group. Taken together, BmZfrp8 may have a critical biological role in of B. mori, since it regulates the expression of the developmental pathway and cell cycle-associated genes.
... Homocysteine (Hcy), a thiol-containing amino acid produced during the metabolism of methionine, has been shown to be associated with many pathological conditions. Hyperhomocysteinemia (HHcy) can aggravate dextran sulfate sodium-induced colitis by stimulating the expression of inflammatory substances [5], impair endothelial cell function by destroying the endothelial barrier, enhance the permeability of endothelial cells, and drive inflammatory reactions [6,7]. Hcy has also been confirmed to be an important intestinalderived uremic toxin [8], and elevated plasma Hcy levels are common in patients with uremia. ...
Article
Full-text available
Background/aims: Previous studies have shown that homocysteine (Hcy) is an important intestinal-derived uremic toxin. However, whether Hcy is involved in the epithelial barrier dysfunction observed in uremia remains unclear. This study aimed to investigate the effect of Hcy on intestinal permeability and intestinal barrier structure and function in adenine-induced uremic rats. Methods: Sprague-Dawley rats were divided into five groups: normal control (group NC), Hcy (group H), uremia (group U), uremia + Hcy (group UH), and uremia + Hcy + VSL#3 (group UHV). Experimental uremia was induced by intragastric adenine administration, and Hcy was injected subcutaneously. The animal models were assessed for renal function and pathological tissue staining. The pathological changes of intestinal tissue were observed by hematoxylin and eosin staining and electron microscopy. The serum and intestinal tissue levels of Hcy, interleukin (IL)-6, tumor necrosis factor (TNF)-α, superoxide dismutase (SOD), and malondialdehyde (MDA) as well as serum endotoxin and intestinal permeability were assessed. The levels of the tight junction proteins claudin-1, occludin, and zonula occludens-1 (ZO-1) were assessed by western blotting. Results: Blood analyses and renal pathology indicated that experimental uremia was induced successfully. Pathological damage to intestinal structure was most obvious in group UH. Serum and tissue Hcy, serum endotoxin, and intestinal permeability were significantly elevated in group UH. The protein levels of claudin-1, occludin, and ZO-1 were decreased to various degrees in group UH compared with groups NC, H, and U. The serum and tissue levels of IL-6, TNF-α, and MDA were significantly increased, while SOD activity was markedly decreased. Supplementation with the probiotic VSL#3 improved these parameters to various degrees and up-regulated the abundance of tight junction proteins, which indicated a role for Hcy in the increase of intestinal permeability and destruction of the epithelial barrier in uremia. Conclusion: Hcy aggravates the increase of intestinal permeability and destruction of the epithelial barrier by stimulating inflammatory and oxidative damage. Probiotic administration can ameliorate this damage by reducing the levels of Hcy-induced inflammation and oxidation.
... Subsequently, MMP-9 interacts with different components of the BBB and leads to disruption of this structure. Second, Hcy acts as an excitatory neurotransmitter for: (i) γ-aminobutyric acid (GABA) receptors A, which leads to increased vascular permeability [55]; and (ii) NMDA receptor [43]. The expression of NMDA receptor is not confident to neurons only. ...
Article
Full-text available
Homocysteine (Hcy) is a sulfur-containing non-proteinogenic amino acid derived in methionine metabolism. The increased level of Hcy in plasma, hyperhomocysteinemia, is considered to be an independent risk factor for cardio and cerebrovascular diseases. However, it is still not clear if Hcy is a marker or a causative agent of diseases. More and more research data suggest that Hcy is an important indicator for overall health status. This review represents the current understanding of molecular mechanism of Hcy metabolism and its link to hyperhomocysteinemia-related pathologies in humans. The aberrant Hcy metabolism could lead to the redox imbalance and oxidative stress resulting in elevated protein, nucleic acid and carbohydrate oxidation and lipoperoxidation, products known to be involved in cytotoxicity. Additionally, we examine the role of Hcy in thiolation of proteins, which results in their molecular and functional modifications. We also highlight the relationship between the imbalance in Hcy metabolism and pathogenesis of diseases, such as cardiovascular diseases, neurological and psychiatric disorders, chronic kidney disease, bone tissue damages, gastrointestinal disorders, cancer, and congenital defects.
... In our experimental settings, the increase in SOD activity following the incubation with Na 2 S was observed only in Ca 2+ -stimulated muscles. This increment might be explained by increased concentration of its substrate O 2 À due to the said ability to generate it, but also by the ability of H 2 S to react with different ROS, superoxide radical anion and hydrogen peroxide [35,36,37] giving false higher activity. It is plausible that metalloproteins, particularly those that contain heme, represent specific targets of HS À since heme proteins coordinating to sulfide ligands in the iron (III) oxidation state could have either specialized (low polarity) environments or allow only limited access to the HS À /S 2À binding site [38]. ...
Article
Background: Our aim was to investigate the effect of methanethiol (CH3SH) on contractility of rat uterus and activities of redox-active enzymes, and to compare them with the effect of sodium sulphide (Na2S), a hydrogen sulphide (H2S/HS(-)) donor. Methods: Uteri were isolated from virgin Wistar rats, divided into six groups, controls (untreated uteri allowed to contract spontaneously and in the presence of Ca(2+)(6mM)), CH3SH treated (spontaneously active and Ca(2+) induced) and Na2S treated (spontaneously active and Ca(2+) induced). Underlying antioxidative enzyme activities (superoxide dismutase--SOD, glutathione peroxidase--GSHPx, glutathione reductase--GR) in CH3SH- or Na2S-treated uteri were compared to controls. Results: Our experiments showed that CH3SH and Na2S provoked reversible relaxation of both spontaneous and Ca(2+)-induced uterine contractions. The dose-response curves differed in shape, and CH3SH curve was shifted to higher concentration compared to H2S/HS(-). The effects of Na2S fitted sigmoid curve, whereas those of CH3SH fitted linearly. CH3SH provoked increased SOD activity and decreased GR activity. However, Na2S (H2S/HS(-)) provoked an increase in SOD activity exclusively in Ca(2+)-stimulated uteri, while the activity of GSHPx was increased in both types of active uteri. Conclusion: Our results imply that CH3SH may have a constructive role in the control of muscle function and metabolism. Observed differences between CH3SH and H2S/HS(-) could be attributed to a larger moiety that is present in CH3SH compared to H2S, but they are more likely to be a consequence of the specific actions of HS(-), in relation to its negative charge.
... Among these pathological changes, vascular endothelial cell injury has been recognized to be the key factor that contributes to the incidence and aggravation of cardiovascular diseases. Hcy may exert its effects of destroying the endothelial barrier, enhancing the permeability of endothelial cells, and driving inflammatory reaction by promoting the biosynthesis of matrix metalloproteinases (MMPs) [4,5]. ...
Article
Full-text available
To investigate the effect of homocysteine (Hcy) on intestinal permeability in rats with TNBS/ethanol-induced colitis and elucidate its mechanism. Sprague-Dawley rats were divided into four groups: normal, normal + Hcy injection, TNBS model, and TNBS model + Hcy injection. Experimental colitis was induced by trinitrobenzene sulfonic acid (TNBS) in 50% ethanol; rats were injected subcutaneously with Hcy from the first day after the induction of experimental colitis on 30 consecutive days. To determine the severity of colitis, the disease activity index (DAI) was evaluated; colon tissues were collected for the detection of the activity of myeloperoxidase (MPO) and the contents of MDA, IL-1β, IL-6, TNF-α, MMP-2, and MMP-9. Intestinal epithelial permeability was assessed with Evans blue (EB) dye. The levels of Hcy in plasma and colon mucosa were measured by high-performance liquid chromatography-fluorescence detection (HPLC-FD). Compared with the normal group, the DAI scoring and MPO activity, contents of MDA, IL-1β, IL-6, TNF-α, MMP-2, MMP-9 in the colon and EB in the small intestine were significantly increased in the TNBS group (P < 0.01). Compared with the TNBS model group, the DAI scoring, plasma and colonic mucosa Hcy levels, MPO activity and contents of MDA, IL-1β, IL-6, TNF-α, MMP-2, MMP-9 in colon and EB in small intestine were significantly increased in the TNBS-induced colitis rats with simultaneous Hcy injection (P < 0.01). Hcy can increase intestinal permeability and aggravate inflammatory damage in rats with TNBS-induced colitis, the underlying mechanisms of which may be attributed to its effects of promoting the expression of MMP-2 and MMP-9, leading to injury of the intestinal barrier.
Article
An elevated level of homocysteine (Hcy) leads to hyperhomocysteinemia (HHcy), which results in vascular dysfunction and pathological conditions identical to stroke symptoms. Hcy increases oxidative stress and leads to increase in blood–brain barrier permeability and leakage. Hydrogen sulfide (H2S) production during the metabolism of Hcy has a cerebroprotective effect, although its effectiveness in Hcy‐induced neurodegeneration and neurovascular permeability is less explored. Therefore, the current study was designed to perceive the neuroprotective effect of exogenous H 2S against HHcy, a cause of neurodegeneration. To test this hypothesis, we used four groups of mice: control, Hcy, control + sodium hydrosulfide hydrate (NaHS), and Hcy + NaHS, and an HHcy mice model in Swiss albino mice by giving a dose of 1.8 g of dl‐Hcy/L in drinking for 8–10 weeks. Mice that have 30 µmol/L Hcy were taken for the study, and a H 2S supplementation of 20 μmol/L was given for 8 weeks to all groups of mice. HHcy results in the rise of the levels of superoxide and nitrite, although a concomitant decrease in the level of superoxide dismutase, catalase, glutathione peroxidase, reduced glutathione, and arginase in oxidative stress and a concomitant decrease in the endogenous level of H 2S. Although H 2S supplementation ameliorated, the effect of HHcy and the levels of H 2S returned to the average level in HHcy animals supplemented with H 2S. Interestingly, H 2S supplementation ameliorated neurovascular remodeling and neurodegeneration. Thus, our study suggested that H 2S could be a beneficial therapeutic candidate for the treatment of Hcy‐associated neurodegeneration, such as stroke and neurovascular disorders. The cerebroprotective role of hydrogen sulfide (H2S) is investigated in a hyperhomocysteinemia mice model. We report the homocysteine and urea cycle enzyme connection in neurodegeneration, which can be reduced by H 2S treatment.
Chapter
Homocysteinemia causes neurological damage through a variety of molecular mechanisms. The sequelae are, therefore, varied, ranging from neural tube defects to epilepsy to apoptosis and dementia. Folate as well as cobalamin deficiency has been implicated in neural tube defects and both have been demonstrated to mediate these abnormalities through homocysteinemia and the ensuant alterations in methylation reactions which modulate gene expressions. In addition, homocysteinemia causes vascular remodelling of the placental vasculature too, resulting in compromised blood flow to the fetus.Vascular remodelling of the cerebral vessels results in ischemic as well as hemorrhagic events. It has been seen that homocysteinemia is involved in the pathophysiology of almost all types of neurological as well as neuropsychiatric disorders, for e.g epilepsy, parkinson’s disease, multiple sclerosis, peripheral neuropathy, Down’s syndrome, schizophrenia, depression, bipolar disorder, etc. Hence, in these situations, it must be ascertained whether or not the patient has high circulating levels of homocysteine, and, if yes, then modulators of homocysteine may be included in their management.
Article
Full-text available
Alcohol is the most socially accepted addictive drug. Alcohol consumption is associated with some health problems such as neurological, cognitive, behavioral deficits, cancer, heart, and liver disease. Mechanisms of alcohol-induced toxicity are presently not yet clear. One of the mechanisms underlying alcohol toxicity has to do with its interaction with amino acid homocysteine (Hcy), which has been linked with brain neurotoxicity. Elevated Hcy impairs with various physiological mechanisms in the body, especially metabolic pathways. Hcy metabolism is predominantly controlled by epigenetic regulation such as DNA methylation, histone modifications, and acetylation. An alteration in these processes leads to epigenetic modification. Therefore, in this review, we summarize the role of Hcy metabolism abnormalities in alcohol-induced toxicity with epigenetic adaptation and their influences on cerebrovascular pathology.
Article
Full-text available
The establishment of hippocampal long-term (LTP) requires protein and mRNA synthesis, suggesting that neuronal activity resulting in LTP initiates a cascade of gene expression. The expression of the gene for the extracellular serine protease tissue plasminogen activator (t-PA) is induced during LTP. Here we analyze long-lasting LTP (L-LTP,>4 hr)in CA1 hippocampal slices of mice homozygous for disrupted t-PA genes. Although mutant mice appear to exhibit long-term potentiation, we provide evidence that these mice are devoid of conventional homosynaptic L-LTP at the Schaffer collateral-CA1 pyramidal cell synapse. Most remarkably, t-PA-deficient mice exhibit a different form of long-lasting potentiation that is characterized by an NMDA receptor- dependent modification of GABA transmission in the CA1 region.
Article
Full-text available
PD 098059 has been shown previously to inhibit the dephosphorylated form of mitogen-activated protein kinase kinase-1 (MAPKK1) and a mutant MAPKK1(S217E,S221E), which has low levels of constitutive activity (Dudley, D. T., Pang, L., Decker, S. J., Bridges, A. J., and Saltiel, A. R.(1995) Proc. Natl. Acad. Sci. U. S. A. 92, 7686-7689). Here we report that PD 098059 does not inhibit Raf-activated MAPKK1 but that it prevents the activation of MAPKK1 by Raf or MEK kinase in vitro at concentrations (IC = 2-7 μM) similar to those concentrations that inhibit dephosphorylated MAPKK1 or MAPKK1(S217E,S221E). PD 098059 inhibited the activation of MAPKK2 by Raf with a much higher IC value (50 μM) and did not inhibit the phosphorylation of other Raf or MEK kinase substrates, indicating that it exerts its effect by binding to the inactive form of MAPKK1. PD 098059 also acts as a specific inhibitor of the activation of MAPKK in Swiss 3T3 cells, suppressing by 80-90% its activation by a variety of agonists. The high degree of specificity of PD 098059 in vitro and in vivo is indicated by its failure to inhibit 18 protein Ser/Thr kinases (including two other MAPKK homologues) in vitro by its failure to inhibit the in vivo activation of MAPKK and MAP kinase homologues that participate in stress and interleukin-1-stimulated kinase cascades in KB and PC12 cells, and by lack of inhibition of the activation of p70 S6 kinase by insulin or epidermal growth factor in Swiss 3T3 cells. PD 098059 (50 μM) inhibited the activation of p42 and isoforms of MAP kinase-activated protein kinase-1 in Swiss 3T3 cells, but the extent of inhibition depended on how potently c-Raf and MAPKK were activated by any particular agonist and demonstrated the enormous amplification potential of this kinase cascade. PD 098059 not only failed to inhibit the activation of Raf by platelet-derived growth factor, serum, insulin, and phorbol esters in Swiss 3T3 cells but actually enhanced Raf activity. The rate of activation of Raf by platelet-derived growth factor was increased 3-fold, and the subsequent inactivation that occurred after 10 min was prevented. These results indicate that the activation of Raf is suppressed and that its inactivation is accelerated by a downstream component(s) of the MAP kinase pathway.
Article
Full-text available
It has been proposed that albumin interacts with the endothelial cell surface to form part of the molecular filter at the capillary wall. Previous investigations of the "protein effect" have been limited to sites accessible to albumin from the capillary lumen. We tested for a specific interaction of albumin with the ablumenal surface of the capillary wall. The hydraulic conductivity (Lp) of single capillaries in frog mesentery, perfused initially with albumin-free Ringer, was reduced when albumin (concentration greater than 1 mg/ml) was added to the superfusate (mean fractional reduction 0.56 +/- 0.05 SE, n = 15). A similar reduction was measured when the mesothelial barrier to water and solute flows between the superfusate and the albuminal surface of the capillary was destroyed. When the albumin was extensively dialyzed against Ringer to remove small vasoactive molecules, no change in the fractional reduction of Lp was observed. Lp was reduced to a minimum value in any capillary when the albumin concentration on both sides of the capillary wall was greater than 1 mg/ml. Our data conform to the hypothesis that albumin modifies structures throughout the capillary wall to maintain normal permeability. We predict that extravascular albumin reduces the ability of a Ringer perfusate to increase permeability in many isolated perfused organs.
Article
Full-text available
We have developed an electrical method to study endothelial cell shape changes in real time in order to examine the mechanisms of alterations in the endothelial barrier function. Endothelial shape changes were quantified by using a monolayer of endothelial cells grown on a small (10(-3) cm2) evaporated gold electrode and measuring the changes in electrical impedance. Bovine pulmonary microvessel endothelial cells and bovine pulmonary artery endothelial cells were used to study the effects of alpha-thrombin on cell-shape dynamics by the impedance measurement. alpha-Thrombin produced a dose-dependent decrease in impedance that occurred within 0.5 min in both cell types, indicative of retraction of endothelial cells and widening of interendothelial junctions because of "rounding up" of the cells. The alpha-thrombin-induced decrease in impedance persisted for approximately 2 hr, after which the value recovered to basal levels. Pretreatment of endothelial cells with the protein kinase C inhibitor, calphostin C, or with 8-bromoadenosine 3',5'-cyclic monophosphate prevented the decreased impedance, suggesting that the endothelial cell change is modulated by activation of second-messenger pathways. The alpha-thrombin-induced decrease in impedance was in agreement with the previously observed increases in transendothelial albumin permeability and evidence of formation of intercellular gaps after alpha-thrombin challenge. The impedance measurement may be a valuable in vitro method for the assessment of mechanisms of decreased endothelial barrier function occurring with inflammatory mediators. Since the rapidly occurring changes in endothelial cell shape in response to mediators such as thrombin are mediated activation of second-messenger pathways, the ability to monitor endothelial cell dynamics in real time may provide insights into the signal-transduction events mediating the increased endothelial permeability.
Article
During early neuronal development, GABA functions as an excitatory neurotransmitter, triggering membrane depolarization, action potentials, and the opening of plasma membrane Ca ²⁺ channels. These excitatory actions of GABA lead to a number of changes in neuronal structure and function. Although the effects of GABA on membrane biophysics during early development have been well documented, little work has been done to examine the possible mechanisms underlying GABA-regulated plastic changes in the developing brain. This study focuses on GABA-regulated kinase activity and transcriptional control. We utilized a combination of Western blotting and immunocytochemical techniques to examine two potential downstream pathways regulated by GABA excitation: the p42/44 mitogen-activated protein kinase (MAPK) cascade and the transcription factor cyclic AMP response element binding protein (CREB). During early development of cultured hypothalamic neurons (5 days in vitro), stimulation with GABA triggered activation of the MAPK cascade and phosphorylation of CREB at Ser 133. These effects were mediated by the GABA A receptor, since administration of the GABA A receptor-specific agonist muscimol (50 μM) triggered pathway activation, and pretreatment with the GABA A -receptor specific antagonist bicuculline (20 μM) blocked pathway activation. Immunocytochemistry revealed a spatial and temporal correlation between activation of the MAPK cascade and CREB phosphorylation. Pretreatment with the MAPK/ERK kinase (MEK) inhibitor U0126 (10 μM) attenuated CREB phosphorylation, indicating that the MAPK pathway regulates that activation state of CREB. In contrast to the excitatory effects observed during early development, in more mature neurons, GABA functions as an inhibitory transmitter. Consistent with this observation, GABA A receptor activation did not stimulate MAPK cascade activation or CREB phosphorylation in mature cultures (18 days in vitro). To determine whether GABA A receptor activation during early development stimulates gene expression, we examined the inducible expression of the neurotrophin brain-derived neurotrophic factor (BDNF). Both GABA and muscimol stimulated BDNF expression, and pretreatment with U0126 attenuated GABA-induced BDNF expression. Whole cell electrophysiological recording was used to assess the effects of BDNF on GABA release. BDNF (100 ng/ml) dramatically increased the frequency of excitatory GABAergic spontaneous postsynaptic currents. Together, these data suggest a positive excitatory feedback loop between GABA and BDNF expression during early development, where GABA facilitates BDNF expression, and BDNF facilitates the synaptic release of GABA. Signaling via the MAPK cascade and the transcription factor CREB appear to play a substantial role in this process.
Article
A protein determination method which involves the binding of Coomassie Brilliant Blue G-250 to protein is described. The binding of the dye to protein causes a shift in the absorption maximum of the dye from 465 to 595 nm, and it is the increase in absorption at 595 nm which is monitored. This assay is very reproducible and rapid with the dye binding process virtually complete in approximately 2 min with good color stability for 1 hr. There is little or no interference from cations such as sodium or potassium nor from carbohydrates such as sucrose. A small amount of color is developed in the presence of strongly alkaline buffering agents, but the assay may be run accurately by the use of proper buffer controls. The only components found to give excessive interfering color in the assay are relatively large amounts of detergents such as sodium dodecyl sulfate, Triton X-100, and commercial glassware detergents. Interference by small amounts of detergent may be eliminated by the use of proper controls.