ArticlePDF Available

Muthalif MM, Benter IF, Karzoun N, Fatima S, Harper J, Uddin MR, Malik KU.20-Hydroxyeicosatetraenoic acid mediates calcium/calmodulin-dependent protein kinase II-induced mitogen-activated protein kinase activation in vascular smooth muscle cells. Proc Natl Acad Sci USA 95:12701-12706

Authors:

Abstract and Figures

Norepinephrine (NE) and angiotensin II (Ang II), by promoting extracellular Ca2+ influx, increase Ca2+/calmodulin-dependent kinase II (CaMKII) activity, leading to activation of mitogen-activated protein kinase (MAPK) and cytosolic phospholipase A2 (cPLA2), resulting in release of arachidonic acid (AA) for prostacyclin synthesis in rabbit vascular smooth muscle cells. However, the mechanism by which CaMKII activates MAPK is unclear. The present study was conducted to determine the contribution of AA and its metabolites as possible mediators of CaMKII-induced MAPK activation by NE, Ang II, and epidermal growth factor (EGF) in vascular smooth muscle cells. NE-, Ang II-, and EGF-stimulated MAPK and cPLA2 were reduced by inhibitors of cytochrome P450 (CYP450) and lipoxygenase but not by cyclooxygenase. NE-, Ang II-, and EGF-induced increases in Ras activity, measured by its translocation to plasma membrane, were abolished by CYP450, lipoxygenase, and farnesyltransferase inhibitors. An AA metabolite of CYP450, 20-hydroxyeicosatetraenoic acid (20-HETE), increased the activities of MAPK and cPLA2 and caused translocation of Ras. These data suggest that activation of MAPK by NE, Ang II, and EGF is mediated by a signaling mechanism involving 20-HETE, which is generated by stimulation of cPLA2 by CaMKII. Activation of Ras/MAPK by 20-HETE amplifies cPLA2 activity and releases additional AA by a positive feedback mechanism. This mechanism of Ras/MAPK activation by 20-HETE may play a central role in the regulation of other cellular signaling molecules involved in cell proliferation and growth.
NE-induced activation of MAPK and cPLA2 but not CaMKII is attenuated by inhibitors of cPLA2, CYP450, and lipoxygenase. Rabbit VSMCs were treated with the vehicle dimethyl sulfoxide (VEH) alone or with inhibitors of cPLA2 (MAFP, 50 M), CaMKII (KN-93, 20 M), or MEK (PD-98059, 20 M) for 4 h or with inhibitors of CYP450 (17-ODYA, 5 M), lipoxygenase (BACL, 5 M), or cyclooxygenase (indomethacin, IND, 5 M) for 30 min and then stimulated with NE (10 M) for 10 min. Cells were harvested, and cell lysates were analyzed. (A) Effect of MAFP on MAPK activity measured by the in-gel MAPK assay. Arrows indicate 44-and 42-kDa bands identified as ERK1 and ERK2. (B) Effect of MAFP on cPLA2, MAPK, and CaMKII activities as measured by in vitro kinase assay. (C-F) NE-induced activation of MAPK and cPLA2 but not CaMKII is attenuated by inhibitors of CYP450 and lipoxygenase but not cyclooxygenase. MAPK activity was determined by in vitro kinase assay (C) and in-gel kinase assay method (D). As a control for protein loading, an unidentified myelin basic protein kinase band is indicated by an arrow. (G) Effect of NE on AA release and cPLA2 activity during blockade of MAPK activity and AA metabolism. Cells were incubated with a combination of 17-ODYA, BACL, and PD-98059 and then stimulated with NE (10 M) for 10 min. Basal values of cPLA2, MAPK, and CaMKII activities and AA release are given in text. The data shown represent the mean SEM of three to six experiments on three batches of cells. , Value significantly different from basal; † , value significantly different from that obtained with VEH of inhibitors; † † , value significantly different from that obtained with either of the inhibitors alone.
… 
Content may be subject to copyright.
Proc. Natl. Acad. Sci. USA
Vol. 95, pp. 12701–12706, October 1998
Pharmacology
20-Hydroxyeicosatetraenoic acid mediates calcium
y
calmodulin-
dependent protein kinase II-induced mitogen-activated protein
kinase activation in vascular smooth muscle cells
M. M. MUTHALIF,I.F.BENTER,N.KARZOUN,S.FATIMA,J.HARPER,M.R.UDDIN, AND K. U. MALIK*
Department of Pharmacology, College of Medicine, The University of Tennessee Center for Health Sciences, Memphis, TN 38163
Communicated by Philip Needleman, Monsanto Company, St. Louis, MO, August 19, 1998 (received for review February 18, 1998)
ABSTRACT Norepinephrine (NE) and angiotensin II (Ang
II), by promoting extracellular Ca
21
influx, increase Ca
21
y
cal-
modulin-dependent kinase II (CaMKII) activity, leading to ac-
tivation of mitogen-activated protein kinase (MAPK) and cyto-
solic phospholipase A
2
(cPLA
2
), resulting in release of arachi-
donic acid (AA) for prostacyclin synthesis in rabbit vascular
smooth muscle cells. However, the mechanism by which CaMKII
activates MAPK is unclear. The present study was conducted to
determine the contribution of AA and its metabolites as possible
mediators of CaMKII-induced MAPK activation by NE, Ang II,
and epidermal growth factor (EGF) in vascular smooth muscle
cells. NE-, Ang II-, and EGF-stimulated MAPK and cPLA
2
were
reduced by inhibitors of cytochrome P450 (CYP450) and lipoxy-
genase but not by cyclooxygenase. NE-, Ang II-, and EGF-induced
increases in Ras activity, measured by its translocation to plasma
membrane, were abolished by CYP450, lipoxygenase, and farne-
syltransferase inhibitors. An AA metabolite of CYP450, 20-
hydroxyeicosatetraenoic acid (20-HETE), increased the activities
of MAPK and cPLA
2
and caused translocation of Ras. These
data suggest that activation of MAPK by NE, Ang II, and EGF
is mediated by a signaling mechanism involving 20-HETE, which
is generated by stimulation of cPLA
2
by CaMKII. Activation of
Ras
y
MAPK by 20-HETE amplifies cPLA
2
activity and releases
additional AA by a positive feedback mechanism. This mecha-
nism of Ras
y
MAPK activation by 20-HETE may play a central
role in the regulation of other cellular signaling molecules
involved in cell proliferation and growth.
Activation of phospholipase A
2
(PLA
2
) liberates arachidonic acid
(AA) from phospholipids. AA metabolites, including prostaglan-
dins, leukotrienes, lipoxins, and hydroxy derivatives, have been
implicated in numerous physiological and pathophysiological
processes (1–5). Recent studies using ‘‘knock-out’’ mice or inhib-
itors indicate that AA-generating cytosolic PLA
2
(cPLA
2
) plays
a role in macrophage production of inflammatory mediators,
reproductive physiology, allergic responses, postischemic brain
injury, cell proliferation, and cancer (6–9). Neurotransmitters,
hormones, and growth factors activate cPLA
2
and protein kinases
in many cell types, including vascular smooth muscle cells
(VSMCs) (10–14). The adrenergic transmitter norepinephrine
(NE) and angiotensin II (Ang II), by promoting extracellular
Ca
21
influx, increases Ca
21
ycalmodulin-dependent kinase II
(CaMKII) activity, leading to activation of mitogen-activated
protein kinase (MAPK) and cPLA
2
, resulting in release of AA for
prostacyclin synthesis (15, 16). This pathway of MAPK activation
by CaMKII is mediated through stimulation of MAPK kinase
(MEK). CaMKIV expressed in PC-12 cells has also been shown
to activate MAPKs (17). MAPKs are also stimulated by
bg
subunits of heterotrimeric G proteins (18, 19). However, the
mechanism by which CaMK activates MAPK is not known.
AA is metabolized by cyclooxygenase into prostaglandins and
thromboxane A
2
, by lipoxygenase into leukotrienes and hydroxy-
eicosatetraenoic acids (HETEs) [5-, 12(S)-, and 15-HETE], and
by cytochrome P450 (CYP450) into epoxyeicosatrienoic acid and
12(R),19- and 20-HETE (1, 3). AA and some of its lipoxygenase
products (12- and 15-HETE) stimulate MAPK activity (20–22).
Moreover, 5-HETE has been shown to increase cPLA
2
activity in
human neutrophils (23). These observations led us to hypothesize
that the NE-induced increase in MAPK activity is caused by AA
or its metabolites generated through activation of cPLA
2
by
CaMKII. To test this hypothesis, we investigated the effects of
NE, Ang II, and epidermal growth factor (EGF) on Ras, MAPK,
CaMKII, and cPLA
2
activity in the presence and absence of
inhibitors of CYP450, lipoxygenase, and cyclooxygenase in rabbit
aortic VSMCs. We have found that NE, Ang II, and EGF activate
the RasyMAPK pathway through generation of a CYP450 me-
tabolite of AA, 20-HETE, after initial activation of cPLA
2
by
CaMKII. Activation of MAPK by 20-HETE amplifies cPLA
2
activity and releases additional AA by a positive feedback mech-
anism.
MATERIALS AND METHODS
Preparation of VSMCs. Aortae were rapidly removed from
male New Zealand White rabbits and the VSMCs were isolated
as described (24). Cells between passages 2 and 8 were plated in
12- or 24-well plates or 100-mm plates. Cells were maintained
under 5% CO
2
in M-199 medium (Sigma) with penicillin, strep-
tomycin, and 10% fetal bovine serum.
Experimental Protocol. VSMCs that were arrested for 48 h
with medium containing 0.05% fetal bovine serum were used for
all studies. Cells were incubated with inhibitors of cPLA
2
(methy-
larachidonylfluorophosphonate, MAFP) (25); MEK (PD-98059)
(26); CaMKII (KN-93) (27); cyclooxygenase (indomethacin)
(28); lipoxygenase (baicalein, BACL) (29); CYP450 (17-
octadecynoic acid, 17-ODYA) (30); farnesyltransferase (FPT III)
(31); or their respective vehicles and exposed to NE (10
m
M); AA
(1–20
m
M); Ang II (100 nM); EGF (100 nM); 5-, 12(R)-, 12(S)-,
15-, or 20-HETEs (1–250 nM); or their vehicles for an additional
5–15 min. The concentrations of various inhibitors used in our
study have been reported to be effective in blocking the activity
of these enzymes in other cell systems (25–31). MAFP and
HETEs were obtained from Cayman Chemicals (Ann Arbor,
MI). NE, myelin basic protein, and indomethacin were from
Sigma. Ang II was from Bachem. AA was from Nu Chek Prep
The publication costs of this article were defrayed in part by page charge
payment. This article must therefore be hereby marked ‘‘advertisement’’ in
accordance with 18 U.S.C. §1734 solely to indicate this fact.
© 1998 by The National Academy of Sciences 0027-8424y98y9512701-6$2.00y0
PNAS is available online at www.pnas.org.
Abbreviations: Ang II, angiotensin II; AA, arachidonic acid; BACL,
baicalein; CaM, calmodulin; CaMKII, Ca
21
yCaM-dependent protein
kinase II; cPLA
2
, cytosolic phospholipase A
2
; CYP450, cytochrome
P450; EGF, epidermal growth factor; HETE, hydroxyeicosatetraenoic
acid; ERK, extracellular regulated kinase; MAFP, methylarachido-
nylfluorophosphonate; MAPK, mitogen-activated protein kinase;
MEK, MAPK kinase; NE, norepinephrine; 17-ODYA, 17-octade-
cynoic acid; VSMC, vascular smooth muscle cell.
*To whom reprint requests should be addressed. e-mail: kmalik@
utmem1.utmem.edu.
12701
(Elysian, MN). PD-98059 was from New England Biolabs. Indo-
methacin, 17-ODYA, and BACL were from Biomol (Plymouth
Meeting, PA). FPT III and KN-93 were from Calbiochem.
Enzyme Assays. In vitro MAPK assays: The activity of MAPK
was determined in VSMC lysates with a Biotrak kit (Amersham)
using a peptide substrate KRELVEPLTPAGEAPNQALLR as
directed by the manufacturer’s instructions.
In-Gel MAPK Assay. MAPK activity was measured in VSMC
homogenates by renaturation assay in polyacrylamide gels con-
taining myelin basic protein as described (32).
In Vitro CaMKII Assay. CaMKII activity was assayed in cell
lysates by using CaMKII assay kits (Upstate Biotechnology) with
a peptide substrate KKALRRQETVDAL by the manufacturer’s
recommendations.
cPLA
2
Assay. cPLA
2
activity in VSMC lysates (20–30
m
gof
protein per assay) was measured by using
L-[1-
14
C]phosphatidyl-
choline (57 mCiymmol, American Radiolabeled Chemicals; 1
Ci 5 37 GBq) as substrate as described (33).
[
3
H]AA Release. VSMCs were labeled with [
3
H]AA (0.25
m
Ciyml; 100 Ciymmol; Dupont-New England Nuclear) for 16 h,
and AA release was measured as described (15).
CYP450 Metabolism of AA in Rabbit VSMCs. VSMC (pas-
sages 2–4), cultured in 150-mm plates, were permeabilized with
Tween 40 (0.1%) for 15 min, washed twice with Krebs buffer, and
incubated with [
14
C]AA (4060 mCiymmol; 0.5
m
Ciyml; NEN)
at 37°C for1hin0.1Mpotassium phosphate (pH 7.4) containing
10 mM MgCl
2
, 1 mM EDTA, 1 mM NADPH, and an NADPH-
regenerating system containing 20 mM isocitrate and 0.1 M
isocitrate dehydrogenase. Cells were incubated with 20
m
MAA
for 5 min and then stimulated with NE (10
m
M) for 30 min. The
aqueous medium was collected, and AA metabolites were ex-
tracted and separated with a binary gradient HPLC system
(Waters Associates) using a Nucleosil C
18
column (5
m
M) with a
two-solvent gradient elution as described (34). The column eluate
was monitored at 235 nm, and radioactivity in the samples was
measured by liquid scintillation spectrometry.
Western Blotting of CYP450 4A. Lysate and microsomal pro-
teins of VSMCs were separated on an SDSypolyacrylamide gel
(10%), transferred to a nitrocellulose membrane, and incubated
FIG. 1. NE-induced activation of MAPK and cPLA
2
but not
CaMKII is attenuated by inhibitors of cPLA
2
, CYP450, and lipoxy-
genase. Rabbit VSMCs were treated with the vehicle dimethyl sulfox-
ide (VEH) alone or with inhibitors of cPLA
2
(MAFP, 50
m
M),
CaMKII (KN-93, 20
m
M), or MEK (PD-98059, 20
m
M)for4horwith
inhibitors of CYP450 (17-ODYA, 5
m
M), lipoxygenase (BACL, 5
m
M),
or cyclooxygenase (indomethacin, IND, 5
m
M) for 30 min and then
stimulated with NE (10
m
M) for 10 min. Cells were harvested, and cell
lysates were analyzed. (A) Effect of MAFP on MAPK activity
measured by the in-gel MAPK assay. Arrows indicate 44- and 42-kDa
bands identified as ERK1 and ERK2. (B) Effect of MAFP on cPLA
2
,
MAPK, and CaMKII activities as measured by in vitro kinase assay.
(C–F) NE-induced activation of MAPK and cPLA
2
but not CaMKII
is attenuated by inhibitors of CYP450 and lipoxygenase but not
cyclooxygenase. MAPK activity was determined by in vitro kinase
assay (C) and in-gel kinase assay method (D). As a control for protein
loading, an unidentified myelin basic protein kinase band is indicated
by an arrow. (G) Effect of NE on AA release and cPLA
2
activity
during blockade of MAPK activity and AA metabolism. Cells were
incubated with a combination of 17-ODYA, BACL, and PD-98059 and
then stimulated with NE (10
m
M) for 10 min. Basal values of cPLA
2
,
MAPK, and CaMKII activities and AA release are given in text. The
data shown represent the mean 6 SEM of three to six experiments on
three batches of cells. p, Value significantly different from basal;
,
value significantly different from that obtained with VEH of inhibi-
tors;
††
, value significantly different from that obtained with either of
the inhibitors alone.
FIG. 2. Production of 20-HETE and expression of CYP450 in
VSMCs. (A) Representative reverse-phase HPLC of products formed
by VSMCs incubated with [
14
C]AA. Solid line, absorbance units full
scale (AUFS); dashed line, radioactivity; arrow, position of authentic
standards. (B) Detection of CYP450 4A in microsomes and lysates of
VSMCs. Approximately 100–200
m
g of proteins from microsomes and
lysates were subjected to SDSyPAGE (10% gel) and detected by
Western blotting using a rat CYP450 4A polyclonal antibody raised in
goats. Lanes (from left to right) are clofibrate-treated liver microsomes
as a standard, VSMC lysates (two lanes), and microsomes isolated
from rabbit VSMCs.
12702 Pharmacology: Muthalif et al. Proc. Natl. Acad. Sci. USA 95 (1998)
for 16 h with rat CYP450 4A antibody (Gentest, Woburn, MA;
1:500 dilution) raised in goats. The immunoblots were developed
by using an enhanced chemiluminescence kit (Amersham).
Raf Phosphorylation. Raf-1 kinase threonine phosphorylation
was determined in cell lysates. The cell lysates were immunopre-
cipitated with anti-phosphothreonine antibody (Sigma). The im-
munoprecipitates were separated in a 7.5% SDSypolyacrylamide
gel and transferred to nitrocellulose membrane; Raf-1 kinase was
detected by the enhanced chemiluminescence method with anti-
Raf-1 kinase antibody (Upstate Biotechnology, Lake Placid,
NY).
Translocation of Ras. Cells were fixed in 4% paraformalde-
hyde in buffer (10 mM Pipesy5 mM EGTAy2 mM MgCl
2
y0.2%
Triton) for 3 min and postfixed in 95% ethanol for 5 min at
220°C. Cells were visualized with anti-Ras (Santa Cruz Biotech-
nology) and Texas red-conjugated horse anti-mouse IgG (Vector
Laboratories) by confocal microscopy (Bio-Rad, MRC-1000,
Laser Scanning Confocal Imaging system using an argonykrypton
lamp) as described (15).
Analysis of Data. The basal values ranged between 916 and
2,387 cpm of [
14
C]AA per 25
m
g of protein per 60 min for cPLA
2
,
11,300 and 26,345 cpm of
32
P per 15
m
g of protein per 10 min for
MAPK, 11,340 and 22,986 cpm of
32
P per 15
m
g of protein per 30
min for CaMKII, and 0.93 and 1.72% fractional AA release in
different batches of cells. The
3
H released into the medium is
expressed as percentage of the total cellular activity and referred
to as fractional AA release. Although the basal values of cPLA
2
,
MAPK, and CaMKII activity and AA release were variable in
different batches of cells, the effect of agonists and inhibitors on
the activity of these enzymes and AA release was consistent
within each batch of cells. Therefore, the changes produced by
agonists and inhibitors have been presented as percent above
basal. The results are expressed as mean 6 SEM. Data were
analyzed with one-way ANOVA. The Newman–Keuls multiple
range test was applied to determine the difference among mul-
tiple groups, and an unpaired Student’s t test was used to
determine the difference between two groups. Differences were
considered significant at P , 0.05.
RESULTS
CYP450 and Lipoxygenase Metabolites of Arachidonic Acid
Mediate NE-Induced MAPK Activity in Rabbit VSMCs. MAFP,
an inhibitor of cPLA
2
(25), attenuated the NE-induced increase
in cPLA
2
and MAPK activity but not in CaMKII activity (Fig. 1
A and B). This raises the possibility that the NE-induced increase
in MAPK activity is caused by AA or its metabolites generated
through activation of cPLA
2
by CaMKII. That an inhibitor of AA
metabolism, 5,8,11,14-eicosatetraynoic acid (35), decreased
MAPK but not CaMKII activity (data not shown) supports this
hypothesis.
If AA metabolites activate MAPK in VSMCs, then inhibition
of the AA-metabolizing enzymes CYP450, lipoxygenase, andyor
cyclooxygenase should attenuate the NE-induced increase in
MAPK and cPLA
2
activities. The inhibitors of CYP450 (17-
ODYA) and, to a lesser degree, lipoxygenase (BACL) attenuated
the NE-induced increase in MAPK and cPLA
2
activity but not
CaMKII activity (Fig. 1CF). Indomethacin did not affect NE-
induced activation of MAPK or cPLA
2
activity. Exogenous AA
increased MAPK and cPLA
2
activity, and this effect was also
inhibited by 17-ODYA and by BACL. AA also increased
CaMKII activity, but this was not inhibited by 17-ODYA or
BACL (data not shown). The inhibitors of CYP450 (17-ODYA),
lipoxygenase (BACL), and MEK (PD-98059) (Fig. 1G), which
diminished the activities of extracellular regulated kinase (ERK)
1 and ERK2 (Fig. 1D and ref. 15), did not completely block the
NE- and AA-induced increases in cPLA
2
activity.
Metabolism of AA by CYP450 and Lipoxygenase in Rabbit
VSMCs. In VSMCs, AA is metabolized by cyclooxygenase to
prostaglandins and thromboxane A
2
and by lipoxygenase to 5-,
12(S)-, or 15-HETE (36). Since the NE-induced increase in
MAPK activity was not only attenuated by a lipoxygenase inhib-
itor but also by a CYP450 inhibitor, we investigated the produc-
tion of CYP450 metabolites of AA in rabbit VSMCs. We found
by reverse-phase HPLC that AA is metabolized by CYP450 to
hydroxy acid(s), one of which has been tentatively identified as
20-HETE (Fig. 2). The calculated amount of this product was
approximately 0.5
m
gymg of protein. Further studies are needed
to confirm the identity of this peak. Moreover, we demonstrated
that CYP4A protein, which may be involved in the formation of
20-HETE (37), is expressed in these cells (Fig. 2).
12(S), 15-, and 20-HETEs Stimulate MAPK and cPLA
2
Ac-
tivities in Rabbit VSMCs. AA has been shown to increase cPLA
2
activity in human neutrophils through formation of 5-HETE (23)
FIG. 3. Effects of 20-HETE and other HETEs on MAPK (A) and cPLA
2
(B) activities and AA release (C) in rabbit VSMCs. VSMCs were
preincubated with 20
m
M PD-98059 (PD) for 4 h and exposed to 20-, 15-, 12(S)-, and 12(R)-HETEs (each at 0.25
m
M) for 10 min. To measure
AA release, VSMCs were labeled with [
3
H]AA for 18 h and then exposed to HETEs for 15 min. The data shown represent the mean 6 SEM of
triplicate measurements of MAPK and cPLA
2
activities and AA release in three batches of cells. Basal values of cPLA
2
, MAPK activity, and AA
release are given in text. p, Value significantly different from basal;
, value significantly different from that obtained with VEH of PD-98059. (D)
Effect of 20-HETE on MAPK activity. Cells were incubated with 20-HETEs (0.25
m
M) for 10 min after pretreatment of cells with or without 5
m
M PD-98059 for4hor5
m
M17-ODYA and 5
m
M BACL for 30 min. This figure is a representation of two experiments.
Pharmacology: Muthalif et al. Proc. Natl. Acad. Sci. USA 95 (1998) 12703
and to increase MAPK activity in rat aortic VSMCs through
formation of 15-HETE (21, 22). In rabbit VSMCs, addition of
12(S)-, 15-, or 20-HETE, but not 12(R)- or 5-HETE (data not
shown), increased MAPK and cPLA
2
activities and AA release
(Fig. 3). 20-HETE also caused activation of MAPK, measured as
ERK1 and ERK2, in the presence of inhibitors of CYP450 and
lipoxygenase (Fig. 3D). 20-HETE increased MAPK activity at
concentrations as low as 0.1
m
M; the maximal effect was pro-
duced at 1
m
M. The effect of 12(S)-, 15-, and 20-HETE of
stimulating MAPK and cPLA
2
was attenuated by the MEK
inhibitor PD-98059 (Fig. 3A), suggesting that these effects could
be mediated by MEK or other signaling elements upstream of
MEK.
20-HETE Mediates NE-Induced Activation of RasyRaf Path-
ways in Rabbit VSMCs. Many agonists stimulate MEK activity by
promoting phosphorylation of proximal kinases, including c-Raf;
Raf is known to be activated by Ras and recruited to the plasma
membrane (38). In the presentstudy, NE and 20-HETE increased
Raf phosphorylation in VSMCs and increased the translocation
of Ras to the plasma membrane. 17-ODYA and BACL blocked
this effect of NE but not of 20-HETE (Fig. 4). Farnesylation of
Ras by farnesyltransferase is required for proper membrane
localization and activity of Ras (39, 40). FPT III, which inhibits
farnesyltransferase activity (31), also blocked the NE- and 20-
HETE-induced translocation of Ras to the plasma membrane
(Fig. 4B). Moreover, FPT III attenuated MAPK and cPLA
2
activities and the AA release elicited by NE (Fig. 4C). Thus,
20-HETE may activate Raf in VSMCs by promoting the trans-
location of Ras to the plasma membrane.
20-HETE Mediates Ang II- and EGF-Induced RasyMAPK
Activation. Ang II and EGF may also stimulate MAPK through
CYP450 and lipoxygenase products of AA released by initial
activation of cPLA
2
by CaMKII. Ang II- and EGF-induced
activation of MAPK was attenuated by inhibitors of CYP450 and
lipoxygenase (Fig. 5A). Moreover, Ang II and EGF caused
translocation of Ras to the plasma membrane, and this was
inhibited by a combination of 17-ODYA and BACL and by FPT
III (Fig. 5B). FPT III also attenuated MAPK activity and AA
release elicited by Ang II and EGF (Fig. 5C). The inhibitory
effect of 17-ODYA and BACL on the Ang II- and EGF-induced
increase in MAPK activity and Ras translocation was reversed by
20-HETE (data not shown).
DISCUSSION
NE, through
a
1
- and
a
2
-adrenergic receptors, stimulates influx of
extracellular Ca
21
(41) and activates CaMKII in rabbit VSMCs
(15). The CaMKII in turn stimulates MAPK and increases cPLA
2
activity, resulting in the release of AA for prostaglandin synthesis
(15). The present study demonstrates a mechanism by which a
CYP450 metabolite of AA, 20-HETE, mediates NE-, Ang II-,
and EGF-stimulated CaMKII-induced activation of the Rasy
MAPK pathway. MAPK then amplifies cPLA
2
activity and
releases further AA for prostaglandin synthesis.
In the present study, the cPLA
2
inhibitor MAFP and the
inhibitor of AA metabolism 5,8,11,14-eicosatetraynoic acid at-
tenuated NE-induced MAPK activity. These results suggest that
a metabolite of AA and not this fatty acid itself is involved in
CaMKII-induced MAPK activation. An important finding is that
inhibitors of CYP450 (17-ODYA) and lipoxygenase (BACL), but
not of cyclooxygenase (indomethacin), attenuated the NE- and
AA-induced increase in MAPK and cPLA
2
and not the CaMKII
activities in VSMCs. From these observations, it follows that
products of AA generated by CYP450 and lipoxygenase but not
by cyclooxygenase contribute to the activation of MAPK and
cPLA
2
elicited by NE. Although the combination of 17-ODYA,
BACL, and the MEK inhibitor PD-98059 abolished the NE-
induced increase in MAPK activity, measured as ERK1 and
ERK2, it only partially reduced cPLA
2
activity. We have reported
that CaMKII mediates activation of cPLA
2
and MAPK in re-
sponse to NE (15). Therefore, it appears that NE-stimulated
CaMKII activates cPLA
2
and releases AA and that products of
AA generated via CYP450 and lipoxygenase stimulate MAPK,
which then amplifies cPLA
2
activity and releases additional AA.
Although CaMKII activates cPLA
2
in VSMCs, it inhibits PLA
2
activity in rat brain synaptosomes (42). This could be due to
differences in the species of PLA
2
; e.g., forskolin enhances cPLA
2
FIG. 4. Involvement of RafyRas in NE-stimulated MAPK path-
way. (A) Effect of NE and 20-HETE on Raf-1 kinase threonine
phosphorylation in VSMCs. VSMCs were pretreated with 17-ODYA
and BACL or vehicle and then stimulated with NE for 5 min, lysed, and
immunoprecipitated with Sepharose-coupled anti-phosphothreonine
antibody. The immunoprecipitates were subjected to SDSyPAGE and
blotted with anti Raf-1 kinase monoclonal antibody and detected by
the enhanced chemiluminescence method. (B) Translocation of Ras in
response to NE (10
m
M) and 20 HETE (0.25
m
M) in the presence of
5
m
M 17-ODYA and 5
m
M BACL (30 min), farnesyltransferase
inhibitor (FPT III, 25
m
M, 24 h), or their vehicle. Cells were visualized
with anti-Ras and Texas red-conjugated horse anti-mouse IgG by
confocal microscopy. This figure is a representation of three experi-
ments. (C) Effect of farnesyltransferase inhibitor (FPT III) on NE-
stimulated MAPK and cPLA
2
activities and AA release. Basal values
of MAPK and cPLA
2
activities and AA release are given in text. The
data shown represent the mean 6 SEM of three to six experiments on
MAPK and cPLA
2
activities and AA release in three batches of cells.
p, Value significantly different from basal;
, value significantly
different from that obtained with VEH of inhibitors.
12704 Pharmacology: Muthalif et al. Proc. Natl. Acad. Sci. USA 95 (1998)
activity in intact synaptosomes but does not alter PLA
2
activity in
rabbit VSMCs (41).
In VSMCs, AA is metabolized by lipoxygenase into 5-, 12-, and
15-HETE (36). The present study indicates that rabbit VSMCs
also express CYP450 4A enzyme, which may be involved in the
formation of 20-HETE. 12(S)-, 15-, and 20-HETE increased
MAPK and cPLA
2
activities and AA release in rabbit VSMCs.
However, 20-HETE (a CYP450 product) was more potent than
12(S)- or 15-HETE (lipoxygenase products) in stimulating
MAPK and cPLA
2
activity. Moreover, the CYP450 inhibitor
(17-ODYA) produced a much greater reduction than the lipoxy-
genase inhibitor (BACL) in NE-induced MAPK and cPLA
2
activities. These results raise the possibility that a product(s) of
AA generated via CYP450, most likely 20-HETE, plays a major
role in regulating NE-induced activation of MAPK and, conse-
quently, amplification of cPLA
2
activity.
Our studies also indicate that MEK mediates NE- and 12(S)-,
15-, and 20-HETE-induced MAPK activation because the MEK
inhibitor PD-98059 attenuated these agents’ effect of increasing
MAPK activity. Since PD-98059 selectively inhibits MEK1 (26),
it appears that MAPK stimulation is mediated primarily by
MEK1. However, we cannot exclude the participation of other
MEK family members that stimulate MAPKs, including c-jun
N-terminal kinase and p38 MAPK.
It is well established that Raf activates MEK (43). Our dem-
onstration that the NE-increased phosphorylation of c-Raf was
inhibited by ODYA and BACL is consistent with a role for Raf
in activation of MEK by NE. Also supporting this view is the
finding that 20-HETE increased Raf phosphorylation in VSMCs.
NE stimulates Ras in human VSMCs (44), and Ras stimulates
Raf by promoting its association with the plasma membrane (45).
The finding that translocation of Ras by NE was blocked by
inhibitors of CYP450 and lipoxygenase suggests that metabolites
of AA activate Raf in VSMCs by promoting the translocation of
Ras to the plasma membrane. The findings that 20-HETE
translocated Ras to the plasma membrane and that the farnesyl-
transferase inhibitor FPT III blocked the Ras translocation
elicited by NE and by 20-HETE supports this contention.
The positive feedback mechanism of RasyMAPK activation by
CYP450 and lipoxygenase products of AA may be a general
mechanism in the actions of hormones like Ang II and growth
factors such as EGF. Ang II and EGF are known to stimulate AA
release, MAPK activity, and cell growth (12, 16, 46, 47). EGF
increased CaMKII activity and AA release in rabbit VSMCs; this
effect was inhibited by removal of calcium from the medium or
addition of the CaMKII inhibitor KN-93 (data not shown). In the
present study, both Ang II and EGF increased MAPK activity
and the translocation of Ras to the plasma membrane, and these
effects were inhibited by 17-ODYA and BACL and by the
farnesyltransferase inhibitor FPT III. It would appear that these
agents, like NE, generate AA metabolites through initial activa-
tion of cPLA
2
by CaMKII, which stimulates Ras and activates
MAPK, amplifies cPLA
2
activity, and releases additional AA for
prostanoid synthesis.
FIG. 5. Ang II and EGF stimulate RasyMAPK pathways through
AA metabolites of CYP450 and lipoxygenase. (A) Effects of 17-
ODYA and BACL on Ang II- and EGF-stimulated MAPK activity.
MAPK activity was measured by in-gel MAPK assay. (B) Transloca-
tion of Ras in response to Ang II and EGF in VSMCs. Arrested
VSMCs were exposed to 5
m
M 17-ODYA and 5
m
M BACL (30 min),
FPT III (24 h, 25
m
M), or their vehicle. Cells were visualized by
confocal microscopy. (C) Effects of FPT III on Ang II- and EGF-
stimulated MAPK activity. Basal values of MAPK are given in text. p,
Value significantly different from basal;
, value significantly different
from that obtained with VEH of FPT III. The data represent the
mean 6 SEM of three to six experiments on three batches of cells.
FIG. 6. Schematic model illustrating 20, 12(S)-, and 15-HETE as
mediators of MAPK and cPLA
2
activation in response to NE, Ang II,
and EGF. In this model, activation of receptors with NE, Ang II, or
EGF leads to an influx of Ca
21
ions that bind to CaM and activate
CaMKII. CaMKII activates cPLA
2
and releases AA. CYP450 and, to
a lesser, extent lipoxygenase metabolites of AA activate MAPK by the
RasyRafyMEK pathway by a positive feedback mechanism. Activation
of MAPK amplifies cPLA
2
activity and further releases AA. R,
receptor; RTK, receptor tyrosine kinase; G, G protein.
Pharmacology: Muthalif et al. Proc. Natl. Acad. Sci. USA 95 (1998) 12705
In rat VSMCs, Ang II-induced MAPK activation has been
reported to be Ras-independent (47, 48). However, another study
conducted in the same cell system proposed that Ang II-induced
MAPK activation was Ras-dependent and is mediated by an
unidentified Ca
21
yCaM-dependent tyrosine kinase through
transactivation of the EGF receptor (14, 49).
The mechanism by which 20-HETE activates Ras is unknown
and is currently being investigated. HETEs may activate Ras by
hydroxy arachidonylation of Ras, which could promote its binding
to the plasma membrane and subsequent activation. The modi-
fication of
a
subunits of G proteins by myristoylation, arachido-
nylation, andyor palmitoylation (50) and of Ras by farnesylation
(39, 40) is known to be required for anchorage to membrane or
interaction with other proteins. In human platelets,
a
subunits of
G proteins (
a
i
,
a
q
,
a
z,
and
a
13
) have been shown to covalently bind
arachidonate and palmitate but not myristate (50). The present
study proposes a signaling mechanism by which NE, Ang II, and
EGF activate the RasyMAPK pathway through generation of an
AA metabolite of CYP450, 20-HETE (Fig. 6). The activation of
MAPK by 20-HETE amplifies cPLA
2
activity and releases ad-
ditional AA by a positive feedback mechanism (Fig. 6). This
mechanism of RasyMAPK activation by 20-HETE might play a
central role in other signaling processes involved in inflammation
and in cell growth, proliferation, and differentiation.
We thank Drs. Alan H. Stephenson and Andrew J. Lonigro (St. Louis
University) for their generous help in HPLC separation of AA metab-
olites, Anne Estes for technical assistance, Dr. Lauren Cagen for scientific
discussions, and Ms. Jin Emerson-Cobb for editing the manuscript. This
work was supported by National Institutes of Health Grant 19134 from
the National Heart, Lung and Blood Institute (to K.U.M.), an American
Heart Association Tennessee Affiliate Award (I.F.B. and M.M.M.), and
a Center for Neuroscience Fellowship (to S.F.).
1. Makita, K., Falck, J. R. & Capdevila, J. H. (1996) FASEB J. 10,
1456–1463.
2. Vane, J. R. & Botting, R. M. (1997) Semin. Arthritis Rheum. 26, 2–10.
3. Needleman, P. & Isakson, P. C. (1997) J. Rheumatol. 24, 6–9.
4. McGiff, J. C., Steinberg, M. & Quilley, J. (1996) Trends Cardiovasc.
Med. 6, 4–9.
5. Brady, H. R. & Serhan, C. N. (1996) Curr. Opin. Nephrol. Hyperten.
5, 20–27.
6. Uozumi, N., Kume, K., Nagase, T., Nakatani, N., Ishii, S., Tashiro, F.,
Komagata, Y., Maki, K., Ikuta, K., Ouchi, Y., et al. (1997) Nature
(London) 390, 618622.
7. Bonventre, J. V., Huang, Z., Taheri, M. R., O’Leary, E., Li, E.,
Moskowitz, M. A. & Sapirstein, A. (1997) Nature (London) 390,
622–625.
8. Anderson, K. M., Roshak, A., Winkler, J. D., McCord, M. & Marshall,
L. A. (1997) J. Biol. Chem. 272, 30504–30511.
9. Heasley, L. E., Thaler, S., Nicks, M., Price, B., Skorecki, K. &
Nemenoff, R. A. (1997) J. Biol. Chem. 272, 14501–14504.
10. Della Rocca, G. J., van Biesen, T., Daaka, Y., Lutrell, D. K., Lutrell,
L. M. & Lefkowitz, R. J. (1997) J. Biol. Chem. 272, 19125–19132.
11. Davis, R. J. (1993) J. Biol. Chem. 268, 14553–14556.
12. Winitz, S., Gupta, S. K., Qian, N. X., Heasley, L. E., Nemenoff, R. A.
& Johnson, G. L. (1994) J. Biol. Chem. 269, 1889–1895.
13. Yang, H., Lu, D., Vinson, G. P. & Raizada, M. A. (1997) J. Neurosci.
17, 1660–1664.
14. Eguchi, S., Matsumoto, T., Motley, E. D., Utsunomiya, H. & Inagami,
T. (1996) J. Biol. Chem. 271, 14169–14175.
15. Muthalif, M. M., Benter, I. F., Uddin, M. R. & Malik, K. U. (1996)
J. Biol. Chem. 271, 30149–30157.
16. Muthalif, M. M., Benter, I. F., Uddin, M. R., Harper, J. & Malik, K. U.
(1998) J. Pharmacol. Exp. Ther. 284, 388–398.
17. Enslen, H., Tokumitsu, H., Stork, P. J., Davis, R. J. & Soderling, T. R.
(1996) Proc. Natl. Acad. Sci. USA 93, 10803–10808.
18. Crespo, P., Xu, N., Simonds, W. F. & Gutkind, J. S. (1994) Nature
(London) 369, 418420.
19. Jelsema, C. L. & Axelrod, J. (1987) Proc. Natl. Acad. Sci. USA 84,
3623–3627.
20. Cui, X. & Douglas, J. G. (1997) Proc. Natl. Acad. Sci. USA 94,
3771–3776.
21. Rao, G. N., Bass, A. S., Glasgow, W. C., Eling, T. E., Runge, M. S. &
Alexander, R. W. (1994) J. Biol. Chem. 269, 32586–32591.
22. Wen, Y., Nadler, J. L., Gonzales, N., Scott, S., Clauser, E. &
Natarajan, R. (1996) Am. J. Physiol. 271, C1212–C1220.
23. Wijkander, J., Flaherty, J. T. O., Nixon, A. B. & Wykle, R. L. (1995)
J. Biol. Chem. 270, 26543–26549.
24. Nebigil, C. & Malik, K. U. (1992) J. Pharmacol. Exp. Ther. 260,
849858.
25. Balsinde, J. & Dennis, E. A. (1996) J. Biol. Chem. 271, 67586765.
26. Dudley, D. T., Pang, L., Decker, S. J., Bridges, A. J. & Saltiel, A.
(1995) Proc. Natl. Acad. Sci. USA 92, 7686–7689.
27. Sumi, M., Kiuchi, K., Ishikawa, T., Ishii, A., Hagiwara, M., Nagatsu,
T. & Hidaka, H. (1991) Biochem. Biophys. Res. Commun. 181,
968–975.
28. Meade, E. A., Smith, W. L. & Dewitt, D. L. (1993) J. Biol. Chem. 268,
66106614.
29. Sekiya, K. & Ohuda, H. (1982) Biochem. Biophys. Res. Commun. 105,
1090–1095.
30. Zou, A. P., Ma, Y.-H., Sui, Z.-H., De Montellano, P. R. O., Clark,
J. E., Masters, B. S. & Roman, R. J. (1994) J. Pharmacol. Exp. Ther.
268, 474481.
31. Leftheris, K., Kline, T., Vite, G. D., Cho, Y. H., Bhide, R. S., Patel,
D. V., Patel, M. M., Schmidt, R. J., Weller, H. N., Andahazy, M. L.,
et al. (1995) J. Med. Chem. 39, 224–236.
32. Huang, X.-C., Richards, E. M. & Sumners, C. (1996) J. Biol. Chem.
271, 15635–15641.
33. Leslie, C. C. (1990) Methods Enzymol. 187, 216–225.
34. Harder, D. R., Gebremedhin, D., Narayanan, J., Jefcoat, C., Falck,
J. R., Campbell, W. B. & Roman, R. (1994) Am. J. Physiol. 266,
H2098–H2107.
35. Chi, E. Y., Henderson, W. A. & Klebanoff, S. J. (1982) Lab. Invest.
47, 579–585.
36. Larrue, J., Rigaud, M., Razaka, G., Daret, D., Demond-Henri, J. &
Bricaud, H. (1983) Biochem. Biophys. Res. Commun. 112, 242–249.
37. Wang, M. H., Stec, D. E., Balazy, M., Mastyugin, V., Yang, C. S.,
Roman, R. J. & Schwartzman, M. L. (1996) Arch. Biochem. Biophys.
336, 240–250.
38. Zhang, X.-F., Settleman, J., Kyriakis, J. M., Takeuchi-Suzuki, E.,
Elledge, S. J., Marshall, M., Bruder, J. T., Rapp, U. R. & Avruch, J.
(1993) Nature (London) 364, 308–313.
39. Zhang, F. L. & Casey, P. J. (1996) Annu. Rev. Biochem. 65, 241–269.
40. Tschantz, W. R., Furfine, E. S. & Casey, P. J. (1997) J. Biol. Chem. 272,
9989–9993.
41. Nebigil, C. & Malik, K. U. (1993) J. Pharmacol. Exp. Ther. 266,
1113–1124.
42. Piomelli, D., Wang, J. K. T., Sihra, T. S., Nairn, A. C., Czernik, A. J.
& Greengard, P. (1989) Proc. Natl. Acad. Sci. USA 86, 85508554.
43. Howe, L. R., Leevers, S. J., Gomez, N., Nakielny, S., Cohen, P. &
Marshall, C. J. (1992) Cell 71, 335–342.
44. Hu, A., Shi, X., Lin, R. Z. & Hoffmann, B. (1996) J. Biol. Chem. 271,
8977–8982.
45. Zhang, X.-F., Settleman, J., Kyriakis, J. M., Takeuchi-Suzuki, E.,
Elledge, S. J., Marshall, M., Bruder, J. T., Rapp, U. R. & Avruch, J.
(1993) Nature (London) 364, 308–313.
46. Peppelenbasch, M. P., Qiu, R. G., de Vries-Smits, A. M., Tertoolen,
L. G., de Laat, S. W., McCormick, F., Hall, A, Symons, M. H. & Bos,
J. L. (1995) Cell 81, 849856.
47. Takahashi, T., Kawahare, Y., Okuda, M., Ueno, H., Takeshita, A. &
Yokoyama, M. (1997) J. Biol. Chem. 272, 16018–16022.
48. Liao, D.-F., Monia, B., Dean, N. & Berk, B. C. (1997) J. Biol. Chem.
272, 61466150.
49. Eguchi, S., Namaguchi, K., Iwasaki, H., Matsumoto, T., Yamakawa,
T., Utsunomiya, H., Motley, E. D., Kawakatsu, H., Owada, K. M.,
Hirata, Y., Marumo, F. & Inagami, T. (1998) J. Biol. Chem. 273,
88908896.
50. Hallak, A., Muszbek, L., Laposata, M., Belmonte, E., Brass, L. F. &
Manning, D. R. (1994) J. Biol. Chem. 269, 4713–4716.
12706 Pharmacology: Muthalif et al. Proc. Natl. Acad. Sci. USA 95 (1998)
... While the molecular mechanisms through which noradrenergic transmission reducing drugs may achieve antidepressant-like effects are not well understood at this time, one possibility is that they selectively dampen certain intracellular signaling pathways after acting upon alpha and beta-adrenergic G protein-coupled receptors. There is already evidence, for example, that NE modulates the Ras/MAPK, PI3K/Akt, JAK/STAT pathways (Muthalif et al., 1998;Yanagawa et al., 2010;Guo et al., 2013;Maity et al., 2020). ...
Article
Full-text available
Major depressive disorder (MDD) remains a significant public health problem worldwide, and revised treatment strategies are therefore urgently needed, including the creation of novel antidepressant compounds or using existing molecular entities in new ways. Etiologic theories of MDD from decades ago have suggested that synaptic deficiencies of monoaminergic neurotransmitters play a causative role in this neuropsychiatric disorder, and that boosting monoamines with drugs such as SSRIs, SNRIs, TCAs, and MAOIs has antidepressant effects and in some individuals can even induce hypomania or mania. While other factors, such as various intracellular molecular pathways and hippocampal neurogenesis, undoubtedly also play a role in MDD, monoaminergic boosting drugs nonetheless have clearly demonstrated antidepressant properties. There is also, however, a body of studies in the preclinical literature suggesting that monoaminergic transmission reducing drugs, including noradrenergic ones, also have antidepressant-like behavioral properties in rodents. Given that there is increasing evidence that the monoamines have u-shaped or Janus-faced dose-response properties, in which a mid-range value is “optimal” in a variety of behavioral and physiological processes, it is plausible that either too much or too little synaptic norepinephrine in key circuits may exacerbate MDD in some individuals. Here we briefly review rodent depression-related behavioral data, focusing on the forced swim test, from three major classes of noradrenergic transmission reducing drugs (alpha2 agonists, beta blockers, alpha1 antagonists), and find much support for the hypothesis that they have antidepressant-like properties. Whether these drugs are antidepressants in human subjects remains to be determined.
... In NSCLC cells, CYP4A/20-HETE increased the tumor growth rate and metastasis (Yu et al., 2011). The signaling mechanisms in CYP4/20-HETE induced cancer progression are mainly about the activation of MAPK pathway (Muthalif et al., 1998), PI 3 K/Akt (Yu et al., 2011), and reactive oxygen species (ROS) (Guo et al., 2007), which was summarized as Figure 2B. ...
Article
Full-text available
Eicosanoids are a class of functionally bioactive lipid mediators derived from the metabolism of long-chain polyunsaturated fatty acids (PUFAs) mediated by multiple enzymes of three main branches, including cyclooxygenases (COXs), lipoxygenases (LOXs), and cytochrome P450s (CYPs). Recently, the role of eicosanoids derived by COXs and LOXs pathways in the control of physiological and pathological processes associated with cancer has been well documented. However, the role of CYPs-mediated eicosanoids, such as epoxyeicosatrienoic acids (EETs), epoxyoctadecenoic acids (EpOMEs), epoxyeicosatetraenoic acids (EpETEs), and epoxydocosapentaenoic acids (EDPs), as well as hydroxyeicosatetraenoic acids (HETEs), in tumorigenesis and cancer progression have not been fully elucidated yet. Here we summarized the association of polymorphisms of CYP monooxygenases with cancers and the pleiotropic functions of CYP monooxygenase-mediated eicosanoids (EETs, EpOMEs, EpETE, EDPs, and 20-HETE) in the tumorigenesis and metastasis of multiple cancers, including but not limited to colon, liver, kidney, breast and prostate cancers, which hopefully provides valuable insights into cancer therapeutics. We believe that manipulation of CYPs with or without supplement of ω-3 PUFAs to regulate eicosanoid profile is a promising strategy to prevent and/or treat cancers.
Article
Coronary reactive hyperemia (CRH) is impaired in cardiovascular diseases, and angiotensin-II (Ang-II) exacerbates it. However, it is unknown how Ang-II affects CRH in Tie2-sEH Tr (human-sEH-overexpressed) versus wild-type (WT) mice. sEH-overexpression resulted in CRH-reduction in Tie2-sEH Tr versus WT. We hypothesized that Ang-II exacerbates CRH-reduction in Tie2-sEH Tr versus WT. Coronary flow in Tie2-sEH Tr and WT was measured by Langendorff system. The hearts were exposed to 15-second ischemia, and CRH was assessed in 10 mice each. Repayment volume was reduced by 40.50% in WT-treated with Ang-II versus WT (7.42 ± 0.8 to 4.49 ± 0.8 ml/g) and 48% in Tie2-sEH Tr-treated with Ang-II versus Tie2-sEH Tr (5.18 ± 0.4 to 2.68 ± 0.3 ml/g). Ang-II decreased repayment duration by 50% in WT-treated with Ang-II versus WT (2.46 ± 0.5 to 1.24 ± 0.4 min) and 54% in Tie2-sEH Tr-treated with Ang-II versus Tie2-sEH Tr (1.66 ± 0.4 to 0.76 ± 0.2 min). Peak repayment flow was reduced by 11.2% in WT-treated with Ang-II versus WT (35.98 ± 0.7 to 32.11 ± 1.4 ml/g) and 4% in Tie2-sEH Tr-treated with Ang-II versus Tie2-sEH Tr (32.18 ± 0.6 to 30.89 ± 1.5 ml/g). Furthermore, CF was reduced by 43% in WT-treated with Ang-II versus WT (14.2 ± 0.5 to 8.15 ± 0.8 ml/min/g) and 32% in Tie2-sEH Tr-treated with Ang-II versus Tie2-sEH Tr (12.1 ± 0.8 to 8.3 ± 1.2 ml/min/g). Moreover, the Ang-II-AT 1 -receptor and CYP4A were increased in Tie2-sEHTr. Our results demonstrate that Ang-II exacerbates CRH-reduction in Tie2-sEH Tr mice.
Article
Blood flow through intrapulmonary arteriovenous anastomoses (QIPAVA) increases during exercise breathing room air but it has been proposed that QIPAVA is reduced during exercise while breathing a fraction of inspired oxygen (FIO2) of 1.00. It has been argued that the reduction in saline contrast bubbles though IPAVA is due to altered in-vivo microbubble dynamics with hyperoxia reducing bubble stability, rather than closure of IPAVA. To definitively determine whether breathing hyperoxia decreases saline contrast bubble stability in vivo, the present study included individuals with and without patent foramen ovale (PFO) to determine if hyperoxia also eliminates left heart contrast in people with an intracardiac right-to-left shunt. Thirty-two participants: 16 without a PFO; 8 female, 8 with a PFO; 4 female, and 8 with late appearing left-sided contrast (4 female) completed five, 4-min bouts of constant-load cycle ergometer exercise (Males - 250 W, Females - 175 W), breathing an FIO2=0.21, 0.40, 0.60, 0.80 and 1.00 in a balanced, Latin Squares design. QIPAVA was assessed at rest and 3-min into each exercise bout via transthoracic saline contrast echocardiography and our previously used bubble scoring system. Bubble scores at FIO2=0.21, 0.40 and 0.60 were unchanged and significantly greater than at FIO2=0.80 and 1.00 in those without a PFO. Participants with a PFO had greater bubble scores at FIO2=1.00 than those without a PFO. These data suggest that hyperoxia-induced decreases in QIPAVA during exercise occur when FIO2≥0.80 and is not a result of altered in vivo microbubble dynamics supporting the idea that hyperoxia closes QIPAVA.
Article
The unsatisfactory rates of adequate blood pressure control among patients receiving antihypertensive treatment calls for new therapeutic strategies to treat hypertension. Several studies have shown that oral sodium nitrite exerts significant antihypertensive effects, but the mechanisms underlying these effects remain unclear. While these mechanisms may involve nitrite-derived S-nitrosothiols, their implication in important alterations associated with hypertension, such as aberrant α1-adrenergic vasoconstriction, has not yet been investigated. Here, we examined the effects of oral nitrite treatment on vascular responses to the α1-adrenergic agonist phenylephrine in two-kidney, one clip (2K1C) hypertensive rats and investigated the potential underlying mechanisms. Our results show that treatment with oral sodium nitrite decreases blood pressure and prevents the increased α1-adrenergic vasoconstriction in 2K1C hypertensive rats. Interestingly, we found that these effects require vascular protein S-nitrosylation, and to investigate the specific S-nitrosylated proteins we performed an unbiased nitrosoproteomic analysis of vascular smooth muscle cells (VSMCs) treated with the nitrosylating compound S-nitrosoglutathione (GSNO). This analysis revealed that GSNO markedly increases the nitrosylation of calcium/calmodulin-dependent protein kinase II γ (CaMKIIγ), a multifunctional protein that mediates the α1-adrenergic receptor signaling. This result was associated with reduced α1-adrenergic receptor-mediated CaMKIIγ activity in VSMCs. We further tested the relevance of these findings in vivo and found that treatment with oral nitrite increases CaMKIIγ S-nitrosylation and blunts the increased CaMKIIγ activity induced by phenylephrine in rat aortas. Collectively, these results are consistent with the idea that oral sodium nitrite treatment increases vascular protein S-nitrosylation, including CaMKIIγ as a target, which may ultimately prevent the increased α1-adrenergic vasoconstriction induced by hypertension. These mechanisms may help to explain the antihypertensive effects of oral nitrite and hold potential implications in the therapy of hypertension and other cardiovascular diseases associated with abnormal α1-adrenergic vasoconstriction.
Article
The incidence of heart failure (HF) is generally preceded by cardiac hypertrophy (CH), which is the enlargement of cardiac myocytes in response to stress. During CH, the metabolism of arachidonic acid (AA), which is present in the cell membrane phospholipids, is modulated. Metabolism of AA gives rise to hydroxyeicosatetraenoic acids (HETEs) and epoxyeicosatrienoic acids (EETs) via cytochrome P450 (CYP) ω-hydroxylases and CYP epoxygenases, respectively. A plethora of studies demonstrated the involvement of CYP-mediated AA metabolites in the pathogenesis of CH. Also, inflammation is known to be a characteristic hallmark of CH. In this review, our aim is to highlight the impact of inflammation on CYP-derived AA metabolites and CH. Inflammation is shown to modulate the expression of various CYP ω-hydroxylases and CYP epoxygenases and their respective metabolites in the heart. In general, HETEs such as 20-HETE and mid-chain HETEs are pro-inflammatory, while EETs are characterized by their anti-inflammatory and cardioprotective properties. Several mechanisms are implicated in inflammation-induced CH, including the modulation of NF-κB and MAPK. This review demonstrated the inflammatory modulation of cardiac CYPs and their metabolites in the context of CH and the anti-inflammatory strategies that can be employed in the treatment of CH and HF.
Article
Objective: 20-Hydroxyeicosatetraenoic acid (20-HETE) is a vasoactive eicosanoid exhibiting effects on vascular smooth muscle cell (VSMC) via G-protein coupled receptor 75 (GPR75) and include stimulation of contractility, migration, and growth. We examined whether VSMC-targeted overexpression of CYP4A12, the primary 20-HETE-producing enzyme in mice, is sufficient to promote hypertension. Methods: Mice with VSM-specific Cyp4a12 overexpression (Myh11-4a12) and their littermate controls (WT) were generated by crossbreeding Cyp4a12-floxed with Myh11-Cre mice. The 20-HETE receptor blocker, N-disodium succinate-20-hydroxyeicosa-6(Z),15(Z)-diencarboxamide (AAA), was administered in the drinking water. Experiments were carried out for 12 days. SBP was measured by tail cuff. Renal interlobar and mesenteric arteries were harvested for assessment of gene expression, 20-HETE levels, vascular contractility, vasodilation, and remodeling. Results: Vascular and circulatory levels of 20-HETE were several folds higher in Myh11-4a12 mice compared with WT. The Myh11-4a12 mice compared with WT were hypertensive (145 ± 2 vs. 127 ± 2 mmHg; P < 0.05) and their vasculature displayed a contractile phenotype exemplified by increased contractility, reduced vasodilatory capacity, and increased media to lumen ratio. All these features were reversed by the administration of AAA. The mechanism of increased contractility includes, at least in part, Rho-kinase activation followed by increased myosin light chain phosphorylation and activation of the contractile apparatus. Conclusion: VSM-specific Cyp4a12 overexpression is sufficient to alter VSM cell phenotype through changes in contractile markers and enhancement in contractility that promote hypertension and vascular dysfunction in a 20-HETE-dependent manner. The 20-HETE receptor GPR75 may represent a novel target for the treatment of hypertension and associated vascular conditions.
Article
Full-text available
Diabetes mellitus is a major debilitating disease whose global incidence is progressively increasing with currently over 463 million adult sufferers and this figure will likely reach over 700 million by the year 2045. It is the complications of diabetes such as cardiovascular, renal, neuronal and ocular dysfunction that lead to increased patient morbidity and mortality. Of these, cardiovascular complications that can result in stroke and cardiomyopathies are 2- to 5-fold more likely in diabetes but the underlying mechanisms involved in their development are not fully understood. Emerging research suggests that members of the Epidermal Growth Factor Receptor (EGFR/ErbB/HER) family of tyrosine kinases can have a dual role in that they are beneficially required for normal development and physiological functioning of the cardiovascular system (CVS) as well as in salvage pathways following acute cardiac ischemia/reperfusion injury but their chronic dysregulation may also be intricately involved in mediating diabetes-induced cardiovascular pathologies. Here we review the evidence for EGFR/ErbB/HER receptors in mediating these dual roles in the CVS and also discuss their potential interplay with the Renin-Angiotensin-Aldosterone System heptapeptide, Angiotensin-(1-7), as well the arachidonic acid metabolite, 20-HETE (20-hydroxy-5, 8, 11, 14-eicosatetraenoic acid). A greater understanding of the multi-faceted roles of EGFR/ErbB/HER family of tyrosine kinases and their interplay with other key modulators of cardiovascular function could facilitate the development of novel therapeutic strategies for treating diabetes-induced cardiovascular complications.
Chapter
Glioblastoma (GBM) is an aggressive form of central nervous system tumors that can occur in the brain or spinal cord. GBM treatment is challenging with limited options with traditional surgery, radiation, and chemotherapy due to mutational heterogeneity, hypervascularity, and predominant hypoxia throughout the tumors. When used as monotherapy or combination therapies, treatments involving angiogenesis pathway-based, mutation-based, or immune-based therapies have shown limited success in decreasing the GBM growth and improving survival, warranting urgent attention in both clinic and laboratory. We have been actively investigating the arachidonic acid (AA) metabolism pathways, where cytochrome P450 4 (CYP4) family of enzymes can produce 20-hydroxyeicosatetraenoic acid (20-HETE) metabolite, a critical signaling eicosanoid contributing to therapeutic resistance. We have shown that 20-HETE can activate several intracellular protein kinases, proinflammatory mediators, and chemokines in GBM. This chapter is focused on understanding the role of the AA metabolic pathway in GBM with an emphasis on the CYP4A-20-HETE axis as a novel therapeutic target. We have also discussed all the effective investigational mechanisms on how 20-HETE can promote GBM refractoriness to available therapies and how its inhibition could sensitize the tumor for improved outcomes.
Article
This article describes the pathways of eicosanoid synthesis, eicosanoid receptors, the action of eicosanoids in different physiological systems, the roles of eicosanoids in selected diseases, and the major inhibitors of eicosanoid synthesis and action. Eicosanoids are oxidised derivatives of 20-carbon polyunsaturated fatty acids (PUFAs) formed by the cyclooxygenase (COX), lipoxygenase (LOX) and cytochrome P450 (cytP450) pathways. Arachidonic acid (ARA) is the usual substrate for eicosanoid synthesis. The COX pathways form prostaglandins (PGs) and thromboxanes (TXs), the LOX pathways form leukotrienes (LTs) and lipoxins (LXs), and the cytP450 pathways form various epoxy, hydroxy and dihydroxy derivatives. Eicosanoids are highly bioactive acting on many cell types through cell membrane G-protein coupled receptors, although some eicosanoids are also ligands for nuclear receptors. Because they are rapidly catabolised, eicosanoids mainly act locally to the site of their production. Many eicosanoids have multiple, sometimes pleiotropic, effects on inflammation and immunity. The most widely studied is PGE2. Many eicosanoids have roles in the regulation of the vascular, renal, gastrointestinal and female reproductive systems. Despite their vital role in physiology, eicosanoids are often associated with disease, including inflammatory disease and cancer. Inhibitors have been developed that interfere with the synthesis or action of various eicosanoids and some of these are used in disease treatment, especially for inflammation.
Article
Full-text available
Mutations in Ras family members that confer oncogenic potential are frequently observed in specific human cancers. We report that human non-small cell lung cancer (NSCLC) lines that harbor oncogenic mutations in Ki-Ras (H460, A549, H2122) synthesized high levels of prostaglandin E2 (PGE2) compared with NSCLC lacking Ras mutations or non-transformed lung epithelial cells (BEAS-2B). This increased PGE2 production was mediated by constitutively high expression of 85-kDa cytosolic phospholipase A2 (cPLA2) and cyclooxygenase 2 (COX-2). The increased expression of cPLA2 protein was mediated through elevated mRNA levels and activation of the cPLA2 promoter. Induction of cPLA2 promoter activity was blocked by expression of dominant-negative forms of Ras. Inhibition of Ras by the farnesyltransferase inhibitor BZA-5B inhibited prostaglandin synthesis in H2122 cells by decreasing expression of both cPLA2 and COX-2. Finally, inhibitors of eicosanoid synthesis blocked anchorage-independent growth of NSCLC lines exhibiting Ki-Ras mutations. These results identify cPLA2 as a novel Ras-inducible regulator of eicosanoid synthesis that participates in cellular transformation.
Article
Full-text available
Receptor-stimulated arachidonic acid (AA) mobilization in P388D macrophages consists of a transient phase in which AA accumulates in the cell and a sustained phase in which AA accumulates in the incubation medium. We have shown previously that a secretory group II phospholipase A (sPLA) is the enzyme responsible for most of the AA released to the incubation medium. By using selective inhibitors for each of the PLAs present in P388D macrophages, we demonstrate herein that the cytosolic group IV PLA (cPLA) mediates accumulation of cell-associated AA during the early steps of P388D cell activation. The contribution of both cPLA and sPLA to AA release can be distinguished on the basis of the different spatial and temporal characteristics of activation and substrate preferences of the two phospholipase As (PLAs). Furthermore, the results suggest the possibility that a functionally active cPLA may be necessary for sPLA to act. cPLA action precedes that of sPLA, and overcoming cPLA inhibition by artificially increasing intracellular free AA levels restores extracellular AA release. Although this suggests cross-talk between cPLA and sPLA, selective inhibition of one other PLA present in these cells, namely the Ca-independent PLA, does not block, but instead enhances receptor-coupled AA release. These data indicate that Ca-independent PLA does not mediate AA mobilization in P388D macrophages. Collectively, the results of this work suggest that each of the PLAs present in P388D macrophages serves a distinct role in cell activation and signal transduction.
Article
Full-text available
We have recently reported that angiotensin II (Ang II)-induced mitogen-activated protein kinase (MAPK) activation is mainly mediated by Ca2+-dependent activation of a protein tyrosine kinase through Gq-coupled Ang II type 1 receptor in cultured rat vascular smooth muscle cells (VSMC). In the present study, we found Ang II rapidly induced the tyrosine phosphorylation of the epidermal growth factor (EGF) receptor and its association with Shc and Grb2. These reactions were inhibited by the EGF receptor kinase inhibitor, AG1478. The Ang II-induced phosphorylation of the EGF receptor was mimicked by a Ca2+ionophore and completely inhibited by an intracellular Ca2+chelator. Thus, AG1478 abolished the MAPK activation induced by Ang II, a Ca2+ ionophore as well as EGF but not by a phorbol ester or platelet-derived growth factor-BB in the VSMC. Moreover, Ang II induced association of EGF receptor with catalytically active c-Src. This reaction was not affected by AG1478. These data indicate that Ang II induces Ca2+-dependent transactivation of the EGF receptor which serves as a scaffold for pre-activated c-Src and for downstream adaptors, leading to MAPK activation in VSMC.
Article
Full-text available
A variety of evidence indicates that activation of Ca2+/calmodulin-dependent protein kinase II (CaM-kinase II) in nerve terminals leads to enhanced neurotransmitter release. Arachidonic acid and its 12-lipoxygenase metabolite, 12-hydroperoxyeicosatetraenoic acid (12-HPETE), have been suggested to act as second messengers mediating presynaptic inhibition of neurotransmitter release. In the present study it was found that CaM-kinase II, purified from rat brain cortex, was inhibited both by arachidonic acid (IC50 = 24 microM) and by 12-HPETE (IC50 = 0.7 microM). Neither substance inhibited CaM-kinase I or III, protein kinase C, or the catalytic subunit of cAMP-dependent protein kinase. Specific inhibition of Ca2+/calmodulin-dependent protein phosphorylation by arachidonic acid was also demonstrated in intact synaptic terminals (synaptosomes) isolated from rat forebrain. These results suggest that arachidonate and its metabolites may modulate synaptic function through the inhibition of CaM-kinase II-dependent protein phosphorylation.
Article
Lipid modifications of proteins can be detected by incubation of cultured cells with radioactive precursors followed by detergent extraction of protein and analysis of the extract by SDS-PAGE and fluorography. Identification of particular proteins requires immunoprecipitation, affinity purification, or differential expression (such as overexpression) before SDS-PAGE analysis. The purpose of this article is to present a brief description of protein acylation and prenylation modifications that have been observed and the details of how to detect the modifications by metabolic radiolabeling of cellular proteins.
Article
Three major enzyme systems-cyclooxygenases, lipoxygenases, and cytochrome P450 monooxygenases (CYP)-generate biological mediators from arachidonic acid (AA). A distinct profile of arachidonate products characterizes each renal tubular segment and each section of the renal vascular tree from arteries to microvessels to veins. The most recent discoveries on renal mechanisms subserved by arachidonate metabolites relate to CYP pathways of AA metabolism. These arachidonate products have prominent effects on blood vessels and ion transport, including modulation and mediation of the actions of vasoactive hormones. The diverse properties of these AA metabolites and the wide distribution of the CYP system make them prime candidates for participation in regulatory mechanisms that affect the circulation and transporting epithelia. CYP-AA products are as important to renal and circulatory control as are nitric oxide and prostaglandins.
Article
Both MAP kinases and the protein kinase p74raf-1 are activated by many growth factors in a c-ras-dependent manner and by oncogenic p21ras. We were therefore interested in determining the relationship between MAP kinases and raf. The MAP kinase ERK2 is activated by expression of oncogenically activated raf, independently of cellular ras. Overexpressed p74raf-1 potentiates activation of ERK2 by EGF and TPA. MAP kinase kinase inactivated by phosphatase 2A treatment is phosphorylated and reactivated by incubation with p74raf-1 immunoprecipitated from phorbol ester-treated cells. We conclude that raf protein kinase is upstream of MAP kinases and is either a MAP kinase kinase kinase or a MAP kinase kinase kinase kinase.
Article
This study was performed to characterize the subtype of adrenergic receptor(s) (AR) involved in prostacyclin synthesis [measured as 6-keto- prostaglandin (PG)F(1α)] elicited by AR agonists in cultured vascular smooth muscle cells of rabbit aorta. Both alpha-1 and alpha-2 AR agonists enhanced 6-keto-PGF(1α) synthesis in a dose-dependent manner with the following order of potency: norepinephrine > BHT 933 > UK 14304 > xylazine > phenylephrine ≥ methoxamine > cirazoline. Isoproterenol and oxymetazoline did not alter 6- keto-PGF(1α) synthesis. Methoxamine-induced 6-keto-PGF(1α) synthesis was not reduced by the alpha-2 AR antagonist rauwolscine. The affinities of AR antagonists (PA2 value) in inhibiting methoxamine-induced 6-keto-PGF(1α) synthesis were of the following order: prazosin > WB 4101 > corynanthine > yohimbine. Administration of WB 4101 and the irreversible alpha-1B AR antagonist chloroethylclonidine reduced norepinephrine (in the presence of rauwolscine, 10-8 M)- or methoxamine-induced 6-keto-PGF(1α) synthesis; WB 4101 was more potent than chloroethylclonidine. UK 14304-induced 6-keto- PGF(1α) synthesis was not reduced by chloroethylclonidine or BRL 44408, a selective alpha-2A AR antagonist, but it was inhibited by other alpha AR antagonists. The affinities of AR antagonists (PA2 values) in inhibiting UK 14304-induced 6-keto-PGF(1α) synthesis were of the following order: rauwolscine > yohimbine > BAM 1303 > BRL 41992 > WB 4101 > ARC 239 ≥ prazosin > SKF 104078 ≥ corynanthine. The order of affinity of alpha-2 AR antagonists in inhibiting UK 14304-induced 6-keto-PGF(1α) synthesis in vascular smooth muscle cells was similar to that derived from radioligand binding studies in opossum kidney cell line receptors classified as alpha-2C receptors. These data suggest that 6-keto-PGF(1α) synthesis elicited by adrenergic stimuli in cultured vascular smooth muscle cells of rabbit aorta is mediated primarily via alpha-2C and to a lesser extent alpha-1A receptors.
Article
We reported that one of the isoquinolinesulfonamide derivatives, KN-62, is a potent and specific inhibitor of Ca2+/calmodulin-dependent protein kinase II (CaMKII) (Tokumitsu, H., Chijiwa, T., Hagiwara, M., Mizutani, A., Terasawa, M. and Hidaka, H. (1990) J. Biol. Chem. 265, 4315-4320). We have now investigated the inhibitory property of a newly synthesized methoxybenzenesulfonamide, KN-93, on CaMKII activity in situ and in vitro. KN-93 elicited potent inhibitory effects on CaMKII phosphorylating activity with an inhibition constant of 0.37 microM but this compound had no significant effects on the catalytic activity of cAMP-dependent protein kinase, Ca2+/phospholipid dependent protein kinase, myosin light chain kinase and Ca(2+)-phosphodiesterase. KN-93 also inhibited the autophosphorylation of both the alpha- and beta-subunits of CaMKII. Kinetic analysis indicated that KN-93 inhibits CaMKII, in a competitive fashion against calmodulin. To evaluate the regulatory role of CaMKII on catecholamine metabolism, we examined the effect of KN-93 on dopamine (DA) levels in PC12h cells. The DA levels decreased in the presence of KN-93. Further, the tyrosine hydroxylase (TH) phosphorylation induced by KCl or acetylcholine was significantly suppressed by KN-93 in PC12h cells while events induced by forskolin or 8-Br-cAMP were not affected. These results suggest that KN-93 inhibits DA formation by modulating the reaction rate of TH to reduce the Ca(2+)-mediated phosphorylation levels of the TH molecule.
Article
The chapter presents a study on macrophage phospholipase A2 specific for sn-2-arachidonic acid. Because most arachidonic acid is esterified at the sn-2 position of phospholipid, phospholipase A2 is thought to play a central role in providing arachidonic acid for subsequent metabolism. In addition, phospholipase A2-mediated hydrolysis of arachidonic acid from the ether-linked membrane phospholipid, 1-O-alkyl-2-arachidonoylglycerophosphocholine (–GPC), is the first step in the production of the potent phospholipid mediator platelet-activating factor (PAF) in inflammatory cells. The lyso-PAF formed is then acetylated to PAF. Compared to diacyl-GPC, the 1-O-alkyl-linked species is enriched in arachidonic acid in neutrophils and macrophages. Consequently, the eicosanoids and PAF can be coordinately produced from a common phospholipid precursor through the initial action of a phospholipase A2. The chapter describes the assay of this arachidonoylspecific phospholipase A2 from the macrophage cell line, RAW 264.7 and the preparation of the 1-O-hexadecyl-2-[3H]arachidonoyl-GPC substrate. The RAW 264.7 macrophage cell line, originally established from a murine tumor induced by Abelson's leukemia virus, secretes lysozyme, is phagocytic, and has receptors for complement and immunoglobulin. In addition, the RAW 264.7 cells release arachidonic acid in response to zymosan and endotoxin.