当前位置: 首页 > 期刊 > 《美国生理学杂志》 > 2005年第4期 > 正文
编号:11295125
AT2 receptors cross talk with AT1 receptors through a nitric oxide- and RhoA-dependent mechanism resulting in decreased phospholipase D acti
http://www.100md.com 《美国生理学杂志》
     Department of Pharmacology, University of Pittsburgh, and Center for Clinical Pharmacology, School of Medicine, Pittsburgh, Pennsylvania

    Department of Pediatrics, Georgetown University Medical Center, Washington, District of Columbia

    ABSTRACT

    ANG II activation of phospholipase D (PLD) is required for ERK and NAD(P)H oxidase activation, both of which are involved in hypertension. Previous findings demonstrate that ANG II stimulates PLD activity through AT1 receptors in a RhoA-dependent mechanism. Additionally, endogenous AT2 receptors in preglomerular smooth muscle cells attenuate ANG II-mediated PLD activity. In the present study, we examined the signal transduction mechanisms used by endogenous AT2 receptors to modulate ANG II-induced PLD activity through either PLA2 generation of lysophosphatidylethanolamine or Gi-mediated generation of nitric oxide (NO) and interaction with RhoA. Blockade of AT2 receptors, Gi and NO synthase, but not PLA2, enhanced ANG II-mediated PLD activity in cells rich in, but not poor in, AT2 receptors. Moreover, NO donors, a direct activator of guanylyl cyclase and a cGMP analog, but not lysophosphatidylethanolamine, inhibited ANG II-mediated PLD activity, whereas an inhibitor of guanylyl cyclase augmented ANG II-induced PLD activity. AT2 receptor- and NO-mediated attenuation of ANG II-induced PLD activity was completely lost in cells transfected with S188A RhoA, which cannot be phosphorylated on serine 188. Therefore, our data indicate that AT2 receptors activate Gi, subsequently stimulating NO synthase and leading to increased soluble guanylyl cyclase activity, generation of cGMP, and activation of a protein kinase, resulting in phosphorylation of RhoA on serine 188. Furthermore, because AT2 receptors inhibit AT1 receptor signaling to PLD via modulating RhoA activity, AT2 receptor signaling can potentially regulate multiple vasoconstrictive signaling systems through inactivating RhoA.

    cGMP; S188a RhoA; phosphorylation of RhoA; hypertension; vascular biology

    EXHAUSTIVE STUDIES HAVE DEMONSTRATED that the spontaneously hypertensive rat (SHR) develops hypertension early in life by a mechanism that is dependent on the renin-angiotensin system, and inhibition of any component of this system normalizes the blood pressure of the SHR to that of its founder line and thus experimental control, the Wistar-Kyoto rat (WKY) (15, 30, 33). Furthermore, the renal vasculature generates the greatest contractile response to ANG II (24). Moreover, extensive studies utilizing kidney transplantation techniques indicate that the blood pressure phenotype is transferred with the kidney, thus indicating that the kidney is the organ responsible for hypertension (19). Not surprisingly, ANG II is a more potent renal vasoconstrictor in SHR than WKY rats (11).

    Therefore, the wealth of data suggests that the primary defect in the SHR resides in ANG II-mediated signal transduction cascades related to contraction in the renal vasculature. In this regard, ANG II, acting via the angiotensin type 1 receptor (AT1R), activates phospholipase D (PLD) more so in SHR compared with WKY preglomerular smooth muscle cells (PGSMCs) (3). PLD, an enzyme that hydrolyzes phosphatidylcholine, generating the lipid second messenger phosphatidic acid (PA), importantly contributes to the vascular effects of ANG II through PA-dependent activation of both the ERK (5) and NAD(P)H oxidase (36).

    Regarding the role of PLD in ANG II-induced activation of the ERK pathway, PA is required for recruitment of Raf-1 to membranes (37). Our studies demonstrate that PLD activity is required for ANG II-mediated activation of MEK in both A10 smooth muscle cells (46) and PGSMCs (4). Therefore, stimulation of PLD activity by ANG II contributes to activation of the Raf-1/MEK/ERK cascade. Because MEK inhibitors, such as PD-98059, attenuate ANG II-mediated smooth muscle contraction (50) and increases in blood pressure (35), PLD activity can lead to increased vascular smooth muscle contraction and ultimately hypertension.

    PLD is also involved in ANG II-mediated activation of NAD(P)H oxidase. In this regard, NAD(P)H oxidase is activated maximally by the synergistic actions of PA and diacylglycerol (36). ANG II generates free radicals via activation of a smooth muscle homolog of NAD(P)H oxidase (28). Moreover, PLD activity is required for ANG II-mediated generation of free radicals in human vascular smooth muscle (51). Furthermore, introduction of free radical scavengers reduces ANG II-induced vasoconstriction (29) and blood pressure in SHR (43). This suggests that PLD-mediated activation of NAD(P)H oxidase contributes to ANG II-induced vasoconstriction and hypertension.

    Because of the important role of PLD in mediating the vascular effects of ANG II, our studies focus on the mechanisms regulating ANG II-mediated PLD activity in vascular smooth muscle cells. Our most recent results indicated that in cultured PGSMCs, the ANG II type 2 receptor (AT2R) attenuates AT1R-induced activation of PLD (6). Thus the overall effect of ANG II on PLD activation depends on the balance of AT1Rs vs. AT2Rs. However, the mechanism by which AT2Rs attenuate AT1R-mediated PLD activation is completely unknown, and the general purpose of the present study was to investigate this mechanism.

    Physiological inhibition of PLD has been demonstrated to occur in plants through a phospholipase A2 (PLA2)-dependent mechanism. Specifically, PLA2 generation of lysophosphatidylethanolamine (LPE) can inhibit PLD through an undefined mechanism (38). Because AT2Rs have been shown to activate PLA (20, 25), AT2R-mediated inhibition of PLD may be PLA and LPE dependent. Alternatively, RhoA, which we showed to be necessary for AT1R-mediated PLD activity (3), can be inactivated through phosphorylation (13). It is well established that phosphorylation of RhoA on serine 188, by cyclic-nucleotide-dependent kinases (PKA and PKG) (13), promotes binding of Rho guanine nucleotide dissociation inhibitors (GDIs) to RhoA thus preventing RhoA from engaging RhoA interacting proteins (16). Moreover, it is known that endogenous AT2R stimulation results in increased levels of renal nitric oxide (NO) and cGMP (47).

    These facts prompted us to examine the hypothesis that AT2Rs inhibit AT1R-mediated PLD activity through a PLA2- and LPE-dependent pathway or a NO- and cGMP-dependent mechanism that involves the inhibitory phosphorylation of RhoA on serine 188. This hypothesis was tested in cultured PGSMCs obtained from WKY rats and, in some experiments, SHR. The rational for using both WKY and SHR PGSMCs was that cultured WKY PGSMCs express sufficient AT2Rs to interfere with AT1R signaling, whereas SHR PGSMCs do not (6).

    METHODS

    Materials. The pharmacological agents used in this work were obtained from the following suppliers: Sigma (St. Louis, MO): ANG II, CGP-42112A, pertussis toxin, and N5-(nitroamidino)-L-2,5-diaminopentanoic acid (NNA); Molecular Probes (Eugene, OR): S-nitroso-N-acetylpenicillamine (SNAP) and diethylamine nitric oxide (DEANO); Alexis (Carlsbad, CA): 1H-[1,2,4]oxadiazole[4,3-a]quinoxalin-1-one (ODQ) and YC-1; Tocris (Ballwin, MO): 4-(4-octadecyl)-4-oxobenzenebutenoic acid (OBAA); Calbiochem (San Diego, CA): G418, 8-Br-cGMP, and botulinum toxin exoenzyme C3. [3H]palmitate was obtained from New England Nuclear/PerkinElmer (Boston, MA). All lipids were obtained from Avanti Polar Lipids (Alabaster, AL), and all molecular biology enzymes were obtained from New England Biolabs (Beverly, MA). PD-123,319 was a gift from Merck. pEF C3 and the vector controls were received from Dr. J. Silvio Gutkind (National Institutes of Health/National Institute of Dental and Craniofacial Research). The S188A RhoA construct was generated as described below.

    Cell culture and PLD assays. Cell culture reagents were obtained from Invitrogen/GIBCO BRL (Carlsbad, CA). PGSMCs were isolated from WKY and SHR from Taconic Farms (Germantown, NY) as described previously (3). The Institutional Animal Care and Use Committee approved all procedures. PLD assays were conducted as described previously (46) and all pharmacological agents were added to the assay media except for DEANO and SNAP, which were added immediately before ANG II. PLD assay data are expressed as the percentage of phosphatidylethanol (PtdEtOH) compared with total radiolabeled lipid. PGSMCs were transfected with Lipofectamine Plus from Invitrogen as per the manufacturer’s directions, and the S188A RhoA-transfected cells were selected for with 200 μg/ml G418.

    Cloning of WKY and SHR RhoA and generation of S188A RhoA. Total RNA was isolated from confluent 100-mm plates using the Rneasy Mini Kit (Qiagen, Valencia, CA). Ten micrograms of total RNA were used in a Clontech Advantage RT-for-PCR Kit (Palo Alto, CA). The resulting cDNA was amplified using Deep Vent polymerase (Invitrogen) with the following primers: 5'-GACGAATTCA-ATGGCTGCCATCAGGAAG-3' and 5'-GGATCCTAC-TGAGGCTGCGTTCACAAG-3'. The product was inserted into the Zero Blunt TOPO PCR Cloning Kit for Sequencing (Invitrogen). The S188A RhoA mutant was generated from WKY RhoA by PCR with the following primers: 5'-TACTCGAGCT-ATGGCTGCCATCAGGAAGAA-3' and 5'-ATCTGCAG-TCACAAGATGAGGCACCCCG(C)CTTTTT-3'. An A-to-C mutation was introduced in the reverse primer at (C) to replace the endogenous serine at position 188 with alanine. The PCR products were ligated into pEGFP-C1 (Clontech), and the resulting colonies were screened and sequenced.

    Data and statistical analysis. The following error propagation formulas were applied. If f and g are two means and fe and ge are their respective error, then the error for f/g is [fe x g – f x ge]/g2 and the error for f ± g is fe+ge. For multiple comparisons, the data were analyzed by ANOVA with Fisher’s least significant difference post hoc test; for individual comparisons, a t-test was used to determine significance. Statistical analysis was conducted using the NCSS 2000 software package (Kaysville, UT). Dose-response curves were analyzed using the curve fit routines of GraphPad Prism (San Diego, CA).

    RESULTS

    NOS and NO mediate AT2R inhibition of AT1R-induced PLD activity. We reported that WKY PGSMCs contain sufficient AT2R expression to interfere with AT1R signaling to PLD and that 10 nM ANG II-mediated PLD activity is enhanced by inhibiting the AT2Rs in WKY but not SHR PGSMCs (6). As shown in Fig. 1, inhibition of NOS with 1 μM NNA, Gi signaling with 100 ng/ml pertussis toxin, and antagonism of AT2Rs with 15 nM PD-123,319 enhanced the ability of 10 nM ANG II-mediated PLD activity to a similar extent in WKY PGSMCs. However, in SHR PGSMCs none of the aforementioned inhibitors significantly enhanced 10 nM ANG II-mediated PLD activity. Inhibition of PLA2, another proposed signal transduction mechanism for the AT2Rs (22, 25), with 700 nM OBAA, used at a concentration shown to inhibit PLA2 (26), had no effect on ANG II-mediated PLD activity in either WKY or SHR PGSMCs. These data indicate that AT2R stimulation of NOS through Gi proteins mediates AT2R inhibition of AT1R-mediated PLD activity in WKY PGSMCs.

    Furthermore, addition of 200 μM concentrations of the NO donors SNAP or DEANO significantly decreased the ability of 1 μM ANG II, a concentration previously shown to give a maximal response in PGSMCs (3), to activate PLD in both WKY and SHR PGSMCs (Fig. 2A). DEA was generated from DEANO by preincubating 200 μM DEANO at 37°C in serum-free media overnight to release all NO from DEANO. The effect of DEA on ANG II-mediated PLD activity was significantly smaller than that of DEANO, indicating that the inhibition from DEANO is due to NO not the donor molecule. The addition of LPE did not inhibit ANG II-mediated PLD activity in WKY or SHR PGSMCs (data not shown), thus confirming that the AT2R is not acting through a PLA- and LPE-dependent mechanism.

    The effect of NO in both WKY and SHR PGSMCs supports our earlier observations that the primary difference between ANG II-mediated PLD regulation is the imbalance of AT1Rs and AT2Rs (6). Moreover, the ANG II dose-response curve generated in the presence of 200 μM DEANO is indistinguishable from the concentration-response curve generated in the presence of 100 nM of the AT2R agonist CGP-42112A (Fig. 2B). These data indicate that the AT2R inhibits AT1R-mediated PLD activity through an NO-dependent mechanism.

    cGMP mediates AT2R inhibition of AT1R-induced PLD activity. The activation of sGC is one the most common modes of NO action. To examine if sGC activation is sufficient to inhibit ANG II-mediated PLD activity, YC-1, a NO-independent activator of sGC (23), was used in WKY PGSMCs. Ten micromolar YC-1 inhibited 1 μM ANG II-mediated PLD activity (Fig. 3). Simultaneous treatment of WKY PGSMCs with 200 μM DEANO and 10 μM YC-1 inhibited ANG II-mediated PLD activity to the same level as YC-1 alone and to a greater extent than DEANO alone, although this latter difference was not statistically significant. Thus the data indicate that NO-mediated sGC activation leads to inhibition of ANG II-mediated PLD activity.

    Ten micromolar sGC inhibitor ODQ (18) potentiated the ability of 10 nM ANG II to stimulate PLD in WKY PGSMCs (Fig. 4A). The magnitude of the effect of ODQ on ANG II-induced PLD activation in WKY PGSMCs was similar to that observed for PD-123,319. Additionally, 100 μM 8-Br-cGMP, a cell-permeable cGMP analog, inhibited 1 μM ANG II-medaited PLD activity in WKY PGSMCs (Fig. 4B). These data confirm that the AT2R inhibits AT1R-mediated PLD activity through a sGC/cGMP-dependent mechanism.

    Signal convergence at RhoA. To confirm our previous findings that RhoA is necessary for ANG II-mediated PLD activity in WKY PGSMCs, we used botulinum toxin exoenzyme C3 that inhibits RhoA activation (2). In WKY PGSMCs scrape loaded with the C3 protein (Fig. 5A) and expressing C3 cDNA (Fig. 5B), 1 μM ANG II failed to activate PLD, whereas 1 μM ANG II activated PLD in control-scraped cells (Fig. 5A) and the vector control (Fig. 5B). Therefore, functional RhoA is required for ANG II-mediated PLD activity in WKY PGSMCs; thus physiological inhibition of RhoA may inhibit ANG II-mediated PLD activity. To test whether AT2R-mediated inhibition of RhoA, through phosphorylation, inhibits ANG II-mediated PLD activity, WKY PGSMCs were stably transfected with S188A RhoA. The S188A RhoA construct generates a mutant RhoA that cannot be phosphorylated and therefore does not interact with GDIs (16). Thus S188A RhoA is only insensitive to the physiological inhibition of RhoA mediated by phosphorylation and subsequent GDI interaction and functions as a wild-type in all other aspects (13).

    In WKY PGSMCs expressing S188A, RhoA ANG II-mediated PLD activity was not enhanced by 15 nM PD-123,319 (Fig. 6A) and was not inhibited by 100 nM CGP-42112A (Fig. 6B). In contrast, PD-123,319 increased and CGP-42112A decreased ANG II-mediated PLD activity in control WKY PGSMCs. Additionally, 200 μM DEANO did not inhibit ANG II-mediated PLD activity in WKY PGSMCs expressing S188A RhoA yet did inhibit ANG II-mediated PLD activity in control cells (Fig. 7A). Furthermore, 200 μM DEANO had no effect on the ANG II-mediated PLD activity concentration-response curve in S188A RhoA-expressing cells (Fig. 7B). As shown in Fig. 7C, the concentration-response curve for ANG II-mediated PLD activity in S188A RhoA cells was shifted to the left and more closely resembled the concentration-response curve of WKY PGSMCs treated with PD-123,319 and that of SHR PGSMCs than control WKY PGSMCs. Consequently, the EC50 values for WKY PGSMCs expressing S188A RhoA are comparable to those of WKY cells treated with PD-123,319 and SHR PGSMCs (Table 1).

    View this table:

    We also compared the sequences of RhoA isolated from WKY and SHR PGSMCs. No differences were found (GenBank accession nos. AY026068 and AY026069). In fact, we found only one difference from the previous rat GenBank entry, D84477, and this apparent mutation is silent.

    DISCUSSION

    AT2Rs appear to participate importantly in vascular biology. In this regard, AT2Rs are present in the vasculature and induce vasorelaxation in vivo (47), and multiple recent reports indicate that the vascular functions of AT2Rs are unmasked when AT1Rs are inhibited (9, 12, 53). Moreover, endogenous AT2Rs are necessary for proper bradykinin-mediated vasodilation (8), and AT2Rs cause NO-induced vasorelaxation (12, 47).

    Data from ANG II receptor knockout mice confirm that there is physiological cross talk, at the level of vascular tone, between AT1Rs and AT2Rs and that AT2Rs are expressed in the adult mouse (1, 48). These data, in conjunction with the pharmacological data, suggest that changes in ANG II receptor levels affect ANG II-mediated signal transduction resulting in alterations in ANG II-mediated blood pressure responses. Our recent findings demonstrate that the ratio of AT1Rs to AT2Rs in cultured WKY PGSMCs is approximately one but is greater in SHR PGSMCs due both to greater expression of AT1Rs and reduced expression of AT2Rs (6). We also found that the imbalance in receptor numbers could account for the increased potency of ANG II, with regard to increasing PLD activity, in SHR PGSMCs. Thus WKY PGSMCs provide a cell culture system for examining endogenous AT2R signaling and SHR PGSMCs provide an unresponsive control group.

    In WKY PGSMCs, blockade of AT2Rs augments ANG II-mediated PLD activity, but this is not seen in SHR PGSMCs (6). These data suggest that AT2Rs suppress ANG II-mediated PLD activity. The results of the present study are consistent with AT2R regulation of AT1R-mediated PLD activation by the following mechanism: Gi-coupled AT2Rs lead to NOS-dependent generation of NO that then stimulates sGC to produce cGMP leading to reduced AT1R-mediated PLD activation through the inhibitory phosphorylation of RhoA.

    Our proposed mechanism for the interaction between AT2Rs and AT1Rs on PLD activity is supported by the results of the current study. In WKY PGSMCs, but not in SHR PGSMCs, blockade of Gi and NOS has similar effects on ANG II-mediated PLD activity as does antagonism of AT2Rs, which supports previous work indicating that AT2Rs are coupled to Gi (21) and can activate NOS (10). Additionally, in WKY PGSMCs NO mimics the effects of stimulating AT2Rs, thus confirming the data obtained with NNA. Due to bypassing AT2Rs, NO also decreases ANG II-mediated PLD activity in SHR PGSMCs. The effect of NO in SHR PGSMCs further confirms our hypothesis that the overriding difference between WKY and SHR PGSMCs with regard to ANG II-mediated PLD activity is the altered ratio of AT1 to AT2 receptors.

    Addition of YC-1 mimics the effects of DEANO, indicating that NO is inhibiting ANG II-mediated PLD activity through sGC. Furthermore, YC-1 and DEANO are not additive suggesting that NO acts only through activation of sGC to inhibit ANG II-mediated PLD activity. Moreover, in WKY PGSMCs, blockade of sGC mimics the effects of antagonism of AT2Rs, thus confirming that AT2R-mediated inhibition of AT1R-mediated PLD activity requires sGC activity. Likewise, a cell-membrane permeant analog of cGMP reduces ANG II-mediated PLD activity, a result consistent with the involvement of sGC in the proposed signal transduction mechanism.

    Consistent with our previous findings, RhoA activity is required for ANG II-mediated PLD activity as demonstrated by the inhibition seen with exotoxin C3. To access the role of phosphorylation of RhoA, we generated a mutant of RhoA that lacks the inhibitory phosphorylation site serine 188, S188A RhoA, and thus cannot be regulated through GDI-dependent mechanisms. In WKY PGSMCs stably expressing S188A RhoA, the ability of PD-123,319 to enhance ANG II-mediated PLD activity is blocked and the ability of CGP-42112A to attenuate ANG II-mediated PLD activity is also blocked. These results indicate a critical role for RhoA in both the signal transduction mechanism for AT1R-medaited activation of PLD and AT2R-mediated inhibition of PLD activity.

    WKY PGSMCs expressing S188A RhoA failed to respond to NO. ANG II concentration-response curves in WKY PGSMCs expressing S188A RhoA more closely resemble SHR and PD-123,319-treated WKY PGSMCs. This supports the notion that AT2R-mediated inhibition of PLD activity is mainly due to the inhibitory actions of the AT2R on RhoA. Moreover, these data indicate that RhoA is the point of negative cross talk between AT1Rs and AT2Rs in regard to PLD activity. Importantly, increased levels of active RhoA have been associated with hypertension in SHR (44, 45). Because no mutations/polymorphisms were found in SHR RhoA, with respect to WKY RhoA, the increased RhoA activity in SHR must arise from aberrant regulation of RhoA.

    RhoA activity is controlled by three mechanisms: 1) activation by guanine nucleotide exchange factors (GEFs), 2) inactivation by GTPase activating proteins (GAPs), and 3) inhibition by GDIs. Increased activity of GEFs can lead to increased RhoA activity; similarly, a decrease in activity of GAPs will lead to prolonged RhoA activation and increased signaling. GDIs interact with guanine-nucleotide-bound RhoA (16), sequester the protein, and relocate RhoA to the cytosol while masking the GEF and effector binding sites (27). Thus AT2R-mediated phosphorylation of RhoA on serine 188 decreases the availability of RhoA for AT1R-mediated activation of PLD, and consequently PLD activity is inhibited. Because the SHR PGSMCs have an imbalance in ANG II receptors that favors AT1Rs (6), RhoA is more active in SHR compared with WKY PGSMCs (TM Seasholtz and EK Jackson, unpublished observations). Therefore, ANG II-mediated RhoA-dependent signaling mechanisms, such as PLD, are increased in SHR compared with WKY (3, 17).

    Taken together, our data support our proposed mechanism by which the AT2R inhibits ANG II-mediated PLD activity. However, the identity of the cyclic nucleotide-dependent kinase involved in the pathway remains unknown. Because cGMP is a known activator of PKG (49) and PKG phosphorylates RhoA at serine 188 (13, 40, 42), the most compelling conclusion is that PKG mediates phosphorylation of RhoA; however, NO/cGMP can lead to the activation of PKA (31, 39, 52), which can also phosphorylate RhoA at the same position (13, 14). Therefore, activation of either kinase could result in the same effect on ANG II-mediated PLD activity.

    Although inhibition of AT2Rs with PD-123,319 and pertussis toxin increases ANG II-induced PLD activation, it is conceivable, although unlikely, that other receptors activated by ANG II mediate, in part, the observed reduction in PLD activation. The experiments presented here were conducted to check each step in the signaling pathway for AT2Rs using an inhibitor and activator of each proposed signaling molecule, including the use of an AT2R antagonist and agonist, which gave opposite results, suggesting that the observed affects can be attributed to the AT2R. However, if there is an alternative ANG II receptor mediating the affects attributed to the AT2R, then the signal transduction cascade by which the alternate receptor interacts with ANG II-mediated PLD activity is the same as we have concluded for the AT2R.

    These data and conclusions may have implications regarding the treatment of hypertension. Angiotensin-converting enzyme (ACE) inhibitors and AT1R blockers (ARBs) are commonly prescribed for patients with hypertension. It is unknown whether it is better to block angiotensin signaling in general via use of ACE inhibitors or just the "pathological signaling" of the AT1Rs with ARBs. ARBs would be expected to attenuate RhoA activity in the renal microcirculation more so than would ACE inhibitors due to the effect on AT2Rs. Because RhoA, through Rho kinases, is involved in smooth muscle contraction, inhibition of RhoA through phosphorylation inhibits contraction (34). Additionally, RhoA activates PLD, which is involved in stimulating NAD(P)H and ERK pathways, therefore inhibiting RhoA may reduce smooth muscle contraction through multiple mechanisms. Moreover, AT2R-mediated inactivation of RhoA may potentially interfere with other activators of RhoA that have been proposed to contribute to hypertension, such as urotensin II (41), endothelin (32), and norepinephrine (7). Therefore, ARBs have a theoretical advantage over ACE inhibitors in that they inhibit the AT1R, consequently inhibiting the vasoconstrictive actions of ANG II while allowing ANG II to stimulate endogenous AT2Rs, thus interfering with general molecular mechanisms of vascular contraction. In addition to avoiding excessive vascular contraction, inhibition of RhoA and consequently PLD should reduce the production of free radicals and cellular proliferation within the renal microcirculation.

    In summary, AT2Rs inhibit AT1R-mediated PLD activation through a NO/cGMP-dependent mechanism most likely mediated by phosphorylation of RhoA at serine 188. Furthermore, this shows that the AT2R does not act as a mere antagonist to AT1R-mediated functions but as an independent receptor with important vascular signaling characteristics. These results imply that selective stimulation of AT2Rs may significantly contribute to the pharmacology of ARBs in the renal microcirculation.

    GRANTS

    B. T. Andresen currently holds a position at Georgetown University Medical School, Washington, D.C. 20007. These studies were funded by National Institutes of Health Grants DK-54782 (to G. G. Romero), DK-02465 (G. G. Romero), and HL-069846 (E. K. Jackson).

    DISCLOSURES

    There are no conflicts of interest or disclosures relevant to these studies with any of the authors.

    ACKNOWLEDGMENTS

    We thank D. G. Gillespie and Zaichuan Mi for assistance in isolation and culturing of the preglomerular smooth muscle cells.

    FOOTNOTES

    The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

    REFERENCES

    Akishita M, Yamada H, Dzau VJ, and Horiuchi M. Increased vasoconstrictor response of the mouse lacking angiotensin II type 2 receptor. Biochem Biophys Res Commun 261: 345–349, 1999.

    Aktories K and Hall A. Botulinum ADP-ribosyltransferase C3: a new tool to study low molecular weight GTP-binding proteins. Trends Pharmacol Sci 10: 415–418, 1989.

    Andresen BT, Jackson EK, and Romero GG. Angiotensin II signaling to phospholipase D in renal microvascular smooth muscle cells in SHR. Hypertension 37: 635–639, 2001.

    Andresen BT, Linnoila JJ, Jackson EK, and Romero GG. Role of EGFR transactivation in angiotensin II signaling to extracellular regulated kinase in preglomerular smooth muscle cells. Hypertension 41: 781–786, 2003.

    Andresen BT, Rizzo MA, Shome K, and Romero G. The role of phosphatidic acid in the regulation of the Ras/MEK/Erk signaling cascade. FEBS Lett 531: 65–68, 2002.

    Andresen BT, Romero GG, and Jackson EK. AT2 receptors attenuate AT1 receptor-induced phospholipase D activation in vascular smooth muscle cells. J Pharmacol Exp Ther 309: 425–431, 2004.

    Asano M and Nomura Y. Comparison of inhibitory effects of Y-27632, a Rho kinase inhibitor, in strips of small and large mesenteric arteries from spontaneously hypertensive and normotensive Wistar-Kyoto rats. Hypertens Res 26: 97–106, 2003.

    Bergaya S, Hilgers RH, Meneton P, Dong Y, Bloch-Faure M, Inagami T, Alhenc-Gelas F, Levy BI, and Boulanger CM. Flow-dependent dilation mediated by endogenous kinins requires angiotensin AT2 receptors. Circ Res 94: 1623–1629, 2004.

    Carey RM, Howell NL, Jin XH, and Siragy HM. Angiotensin type 2 receptor-mediated hypotension in angiotensin type-1 receptor-blocked rats. Hypertension 38: 1272–1277, 2001.

    Carey RM, Jin X, Wang Z, and Siragy HM. Nitric oxide: a physiological mediator of the type 2 (AT2) angiotensin receptor. Acta Physiol Scand 168: 65–71, 2000.

    Chatziantoniou C, Daniels FH, and Arendshorst WJ. Exaggerated renal vascular reactivity to angiotensin and thromboxane in young genetically hypertensive rats. Am J Physiol Renal Fluid Electrolyte Physiol 259: F372–F382, 1990.

    De Godoy MA, De Oliveira AM, and Rattan S. Angiotensin II-induced relaxation of anococcygeus smooth muscle via desensitization of AT1 receptor, and AT2-receptor-mediated relaxation by NOS pathway. J Pharmacol Exp Ther 311: 394–401, 2004.

    Ellerbroek SM, Wennerberg K, and Burridge K. Serine phosphorylation negatively regulates RhoA in vivo. J Biol Chem 278: 19023–19031, 2003.

    Essler M, Staddon JM, Weber PC, and Aepfelbacher M. Cyclic AMP blocks bacterial lipopolysaccharide-induced myosin light chain phosphorylation in endothelial cells through inhibition of Rho/Rho kinase signaling. J Immunol 164: 6543–6549, 2000.

    Ferrone RA and Antonaccio MJ. Prevention of the development of spontaneous hypertension in rats by captopril (SQ 14,225). Eur J Pharmacol 60: 131–137, 1979.

    Forget MA, Desrosiers RR, Gingras D, and Beliveau R. Phosphorylation states of Cdc42 and RhoA regulate their interactions with Rho GDP dissociation inhibitor and their extraction from biological membranes. Biochem J 361: 243–254, 2002.

    Freeman EJ, Ferrario CM, and Tallant EA. Angiotensins differentially activate phospholipase D in vascular smooth muscle cells from spontaneously hypertensive and Wistar-Kyoto rats. Am J Hypertens 8: 1105–1111, 1995.

    Garthwaite J, Southam E, Boulton CL, Nielsen EB, Schmidt K, and Mayer B. Potent and selective inhibition of nitric oxide-sensitive guanylyl cyclase by 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one. Mol Pharmacol 48: 184–188, 1995.

    Grisk O and Rettig R. Renal transplantation studies in genetic hypertension. News Physiol Sci 16: 262–265, 2001.

    Haithcock D, Jiao H, Cui XL, Hopfer U, and Douglas JG. Renal proximal tubular AT2 receptor: signaling and transport. J Am Soc Nephrol 10, Suppl 11: S69–S74, 1999.

    Hansen JL, Servant G, Baranski TJ, Fujita T, Iiri T, and Sheikh SP. Functional reconstitution of the angiotensin II type 2 receptor and Gi activation. Circ Res 87: 753–759, 2000.

    Hiyoshi H, Yayama K, Takano M, and Okamoto H. Stimulation of cyclic GMP production via AT2 and B2 receptors in the pressure-overloaded aorta after banding. Hypertension 43: 1258–1263, 2004.

    Hwang TL, Wu CC, and Teng CM. YC-1 potentiates nitric oxide-induced relaxation in guinea-pig trachea. Br J Pharmacol 128: 577–584, 1999.

    Jackson EK and Herzer WA. Regional vascular selectivity of angiotensin II. J Pharmacol Exp Ther 297: 736–745, 2001.

    Jacobs LS and Douglas JG. Angiotensin II type 2 receptor subtype mediates phospholipase A2- dependent signaling in rabbit proximal tubular epithelial cells. Hypertension 28: 663–668, 1996.

    Kohler T, Heinisch M, Kirchner M, Peinhardt G, Hirschelmann R, and Nuhn P. Phospholipase A2 inhibition by alkylbenzoylacrylic acids. Biochem Pharmacol 44: 805–813, 1992.

    Lang P, Gesbert F, Delespine-Carmagnat M, Stancou R, Pouchelet M, and Bertoglio J. Protein kinase A phosphorylation of RhoA mediates the morphological and functional effects of cyclic AMP in cytotoxic lymphocytes. EMBO J 15: 510–519, 1996.

    Lassegue B, Sorescu D, Szocs K, Yin Q, Akers M, Zhang Y, Grant SL, Lambeth JD, and Griendling KK. Novel gp91(phox) homologues in vascular smooth muscle cells: nox1 mediates angiotensin II-induced superoxide formation and redox-sensitive signaling pathways. Circ Res 88: 888–894, 2001.

    Laursen JB, Rajagopalan S, Galis Z, Tarpey M, Freeman BA, and Harrison DG. Role of superoxide in angiotensin II-induced but not catecholamine-induced hypertension. Circulation 95: 588–593, 1997.

    Madeddu P, Anania V, Varoni MV, Parpaglia PP, Demontis MP, Fattaccio MC, Palomba D, Pollock D, and Glorioso N. Prevention by blockade of angiotensin subtype1-receptors of the development of genetic hypertension but not its heritability. Br J Pharmacol 115: 557–562, 1995.

    Meacci E, Taira M, Moos M Jr, Smith CJ, Movsesian MA, Degerman E, Belfrage P, and Manganiello V. Molecular cloning and expression of human myocardial cGMP-inhibited cAMP phosphodiesterase. Proc Natl Acad Sci USA 89: 3721–3725, 1992.

    Miao L, Dai Y, and Zhang J. Mechanism of RhoA/Rho kinase activation in endothelin-1- induced contraction in rabbit basilar artery. Am J Physiol Heart Circ Physiol 283: H983–H989, 2002.

    Michel JB, Sayah S, Guettier C, Nussberger J, Philippe M, Gonzalez MF, Carelli C, Galen FX, Menard J, and Corvol P. Physiological and immunopathological consequences of active immunization of spontaneously hypertensive and normotensive rats against murine renin. Circulation 81: 1899–1910, 1990.

    Murthy KS, Zhou H, Grider JR, and Makhlouf GM. Inhibition of sustained smooth muscle contraction by PKA and PKG preferentially mediated by phosphorylation of RhoA. Am J Physiol Gastrointest Liver Physiol 284: G1006–G1016, 2003.

    Muthalif MM, Karzoun NA, Gaber L, Khandekar Z, Benter IF, Saeed AE, Parmentier JH, Estes A, and Malik KU. Angiotensin II-induced hypertension: contribution of Ras GTPase/mitogen-activated protein kinase and cytochrome P450 metabolites. Hypertension 36: 604–609, 2000.

    Palicz A, Foubert TR, Jesaitis AJ, Marodi L, and McPhail LC. Phosphatidic acid and diacylglycerol directly activate NADPH oxidase by interacting with enzyme components. J Biol Chem 276: 3090–3097, 2001.

    Rizzo MA, Shome K, Vasudevan C, Stolz DB, Sung TC, Frohman MA, Watkins SC, and Romero G. Phospholipase D and its product, phosphatidic acid, mediate agonist- dependent raf-1 translocation to the plasma membrane and the activation of the mitogen-activated protein kinase pathway. J Biol Chem 274: 1131–1139, 1999.

    Ryu SB, Karlsson BH, Ozgen M, and Palta JP. Inhibition of phospholipase D by lysophosphatidylethanolamine, a lipid- derived senescence retardant. Proc Natl Acad Sci USA 94: 12717–12721, 1997.

    Sandau KB, Gantner F, and Brune B. Nitric oxide-induced F-actin disassembly is mediated via cGMP, cAMP, and protein kinase A activation in rat mesangial cells. Exp Cell Res 271: 329–336, 2001.

    Sauzeau V, Le Jeune H, Cario-Toumaniantz C, Smolenski A, Lohmann SM, Bertoglio J, Chardin P, Pacaud P, and Loirand G. Cyclic GMP-dependent protein kinase signaling pathway inhibits RhoA- induced Ca2+ sensitization of contraction in vascular smooth muscle. J Biol Chem 275: 21722–21729, 2000.

    Sauzeau V, Le Mellionnec E, Bertoglio J, Scalbert E, Pacaud P, and Loirand G. Human urotensin II-induced contraction and arterial smooth muscle cell proliferation are mediated by RhoA and Rho-kinase. Circ Res 88: 1102–1104, 2001.

    Sawada N, Itoh H, Yamashita J, Doi K, Inoue M, Masatsugu K, Fukunaga Y, Sakaguchi S, Sone M, Yamahara K, Yurugi T, and Nakao K. cGMP-dependent protein kinase phosphorylates and inactivates RhoA. Biochem Biophys Res Commun 280: 798–805, 2001.

    Schnackenberg CG, Welch WJ, and Wilcox CS. Normalization of blood pressure and renal vascular resistance in SHR with a membrane-permeable superoxide dismutase mimetic: role of nitric oxide. Hypertension 32: 59–64, 1998.

    Seasholtz TM, Zhang T, Morissette MR, Howes AL, Yang AH, and Brown JH. Increased expression and activity of RhoA are associated with increased DNA synthesis and reduced p27(Kip1) expression in the vasculature of hypertensive rats. Circ Res 89: 488–495, 2001.

    Seko T, Ito M, Kureishi Y, Okamoto R, Moriki N, Onishi K, Isaka N, Hartshorne DJ, and Nakano T. Activation of RhoA and inhibition of myosin phosphatase as important components in hypertension in vascular smooth muscle. Circ Res 92: 411–418, 2003.

    Shome K, Rizzo MA, Vasudevan C, Andresen B, and Romero G. The activation of phospholipase D by endothelin-1, angiotensin II, and platelet-derived growth factor in vascular smooth muscle A10 cells is mediated by small G proteins of the ADP-ribosylation factor family. Endocrinology 141: 2200–2208, 2000.

    Siragy HM and Carey RM. The subtype 2 (AT2) angiotensin receptor mediates renal production of nitric oxide in conscious rats. J Clin Invest 100: 264–269, 1997.

    Siragy HM, Inagami T, Ichiki T, and Carey RM. Sustained hypersensitivity to angiotensin II and its mechanism in mice lacking the subtype-2 (AT2) angiotensin receptor. Proc Natl Acad Sci USA 96: 6506–6510, 1999.

    Smolenski A, Burkhardt AM, Eigenthaler M, Butt E, Gambaryan S, Lohmann SM, and Walter U. Functional analysis of cGMP-dependent protein kinases I and II as mediators of NO/cGMP effects. Naunyn Schmiedebergs Arch Pharmacol 358: 134–139, 1998.

    Touyz RM, El Mabrouk M, He G, Wu XH, and Schiffrin EL. Mitogen-activated protein/extracellular signal-regulated kinase inhibition attenuates angiotensin II-mediated signaling and contraction in spontaneously hypertensive rat vascular smooth muscle cells. Circ Res 84: 505–515, 1999.

    Touyz RM and Schiffrin EL. ANG II-stimulated superoxide production is mediated via phospholipase D in human vascular smooth muscle cells. Hypertension 34: 976–982, 1999.

    Tsukada S, Keino-Masu K, Masu M, and Fukuda J. Activation of protein kinase A by nitric oxide in cultured dorsal root ganglion neurites of the rat, examined by a fluorescence probe, ARII. Neurosci Lett 318: 17–20, 2002.

    Widdop RE, Matrougui K, Levy BI, and Henrion D. AT2 receptor-mediated relaxation is preserved after long-term AT1 receptor blockade. Hypertension 40: 516–520, 2002.(Bradley T. Andresen, Kunt)