ACCELERATED PUBLICATION
Cyclic AMP-independent Activation of Protein Kinase A by Vasoactive Peptides*

Nickolai O. DulinDagger, Jiaxin Niu, Darren D. Browning, Richard D. Ye, and Tatyana Voyno-Yasenetskaya

From the Department of Pharmacology, University of Illinois at Chicago College of Medicine, Chicago, Illinois 60612

Received for publication, April 16, 2001, and in revised form, April 25, 2001


    ABSTRACT
TOP
ABSTRACT
INTRODUCTION
MATERIALS AND METHODS
RESULTS
DISCUSSION
REFERENCES

Protein kinase A (PKA) is an important effector enzyme commonly activated by cAMP. The present study focuses on our finding that the vasoactive peptide endothelin-1 (ET1), whose signaling is not coupled to cAMP production, stimulates PKA in two independent cellular models. Using an in vivo assay for PKA activity, we found that ET1 stimulated PKA in HeLa cells overexpressing ET1 receptors and in aortic smooth muscle cells expressing endogenous levels of ET1 receptors. In these cell models, ET1 did not stimulate cAMP production, indicating a novel mechanism for PKA activation. The ET1-induced activation of PKA was found to be dependent on the degradation of inhibitor of kappa B, which was previously reported to bind and inhibit PKA. ET1 potently stimulated the nuclear factor-kappa B pathway, and this effect was inhibited by overexpression of the inhibitor of kappa B dominant negative mutant (Ikappa Balpha m) and by treatment with the proteasome inhibitor MG-132. Importantly, Ikappa Balpha m and MG-132 had similar inhibitory effects on ET1-induced activation of PKA without affecting Gs-mediated activation of PKA or ET1-induced phosphorylation of mitogen-activated protein kinase. Finally, another vasoactive peptide, angiotensin II, also stimulated PKA in a cAMP-independent manner in aortic smooth muscle cells. These findings suggest that cAMP-independent activation of PKA might be a general response to vasoactive peptides.


    INTRODUCTION
TOP
ABSTRACT
INTRODUCTION
MATERIALS AND METHODS
RESULTS
DISCUSSION
REFERENCES

Endothelin-1 (ET1)1 is a vasoactive peptide implicated in embryonic development and in pathophysiology of cardiovascular, renal, and respiratory systems (1, 2). Two types of ET1 receptors, namely ETA and ETB, have been cloned and identified as typical G protein-coupled receptors (3, 4). ETA receptors are coupled to Gq/11, G12/13, and Gi heterotrimeric G proteins, leading to stimulation of phospholipase C, small GTPase RhoA, and inhibition of adenylyl cyclase, respectively (5-8). The coupling of ET1 receptors to Gs is controversial. A modest cAMP response to ET1 was reported by some investigators (9-11), whereas no response or inhibition of cAMP levels was shown by others (5, 7, 12-15). Moreover, there was no convincing evidence that the main target of cAMP, the protein kinase A (PKA), could be activated by ET1.

The PKA holoenzyme is a tetrameric complex consisting of two catalytic subunits (PKAc) bound to a homodimer of two regulatory subunits (PKAr). The established mechanism of PKA activation in response to various hormones involves stimulatory G proteins, Gs, which activate adenylyl cyclase resulting in production of cAMP. Binding of cAMP to PKAr leads to a release and activation of PKAc (16, 17). Recently, a novel mechanism for PKA activation by lipopolysaccharide (LPS) has been described that is related to the nuclear factor-kappa B (NFkappa B) pathway (18). NFkappa B is a transcription factor that is commonly activated during immune and inflammatory responses (19, 20). Under basal conditions, NFkappa B exists in an inactive state bound to its natural inhibitor Ikappa B. Activation of NFkappa B occurs as a result of agonist-induced phosphorylation and degradation of Ikappa B followed by a release of free NFkappa B. Apparently, a certain pool of PKAc also exists in a complex with Ikappa B (18). Under basal conditions, Ikappa B retains PKAc in the inactive state, presumably by masking its ATP binding site. LPS-induced phosphorylation and degradation of Ikappa B results in a release and activation of PKAc (18). However, except for bacterially derived LPS, there was no evidence that other physiological agonists are able to activate PKA by this mechanism. The present study demonstrates for the first time that ET1 stimulates PKA activity by a cAMP-independent mechanism involving degradation of Ikappa B. Moreover, our data suggest that this is most likely a general phenomenon common for vasoactive peptides.

    MATERIALS AND METHODS
TOP
ABSTRACT
INTRODUCTION
MATERIALS AND METHODS
RESULTS
DISCUSSION
REFERENCES

Reagents-- The cDNA for ETA receptor was kindly provided by Dr. Masashi Yanagisawa (University of Texas, South Western Medical Center, Dallas, TX). The cDNA for FLAG-tagged vasodilator-stimulated phosphoprotein (VASP) was a gift from Dr. Michael Uhler (University of Michigan, Ann Arbor, MI). The cDNA for the dominant negative mutant of PKA (delta R1alpha ) was a gift from Dr. Stanley McKnight (University of Washington, Seattle, WA). The cDNA for the phosphorylation-deficient S32A,S36A mutant of mouse Ikappa Balpha (Ikappa Balpha m) was a gift from Dr. Inder Verma (The Salk Institute, La Jolla, CA). The phosphorylation-deficient S19A,S23A mutant of mouse Ikappa Bbeta (Ikappa Bbeta m) was generated by polymerase chain reaction, and its identity was confirmed by sequencing. The NFkappa B-driven luciferase reporter plasmid was described previously (21). Endothelin-1, isoproterenol, tumor necrosis factor alpha , and MG-132 were from Calbiochem. Angiotensin II was from Peninsula Laboratories. Monoclonal anti-FLAG antibodies were from Sigma. Polyclonal anti-phospho-MAP kinase antibodies were from New England Biolabs.

Cell Culture and DNA Transfection-- The HeLa cells (ATCC) were maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented with 2 mM glutamine, 100 units/ml streptomycin, 100 units/ml penicillin, and 10% fetal bovine serum (FBS). The primary culture of rat aortic smooth muscle cells (RASMC) from Wistar-Kyoto rats was kindly provided by Dr. Sergei Orlov (University of Montreal, Montreal, Canada). The RASMC were cultured for up to 10 passages in DMEM supplemented with 10% FBS, 2 mM glutamine, 100 units/ml streptomycin, and 100 units/ml penicillin as described elsewhere (22). For transient overexpression of proteins, the HeLa cells or RASMC were transfected with desired DNA in the presence of serum, using LipofectAMINE-2000 or LipofectAMINE-Plus reagents (Life Technologies, Inc.), respectively, following the manufacturer's protocol. The cells were serum-starved in 0.2% FBS for 24 h before the experiment.

PKA Activity in Intact Cells-- Phosphorylation-induced electrophoretic mobility shift of the VASP is a highly sensitive functional assay for the activity of cyclic nucleotide-dependent protein kinases in intact cells (23, 24) and was used in this study. The specificity of PKA-mediated phosphorylation of VASP was confirmed by overexpression of the dominant negative mutant of PKA, delta R1alpha , which abolished VASP phosphorylation induced by isoproterenol (see Fig. 1C) or by 8-bromo-cAMP (25) but not by 8-bromo-cGMP (25). The assay involved transient transfection of cells with FLAG-tagged VASP cDNA, stimulation of quiescent cells with desired agonists, cell lysis followed by immunoblotting of cell lysates with FLAG antibodies (see below), and monitoring the phosphorylation-dependent electrophoretic mobility shift of VASP, as described previously (25).

Immunoblotting-- After stimulation of quiescent cells with desired agonists, the cells were lysed in the buffer containing 25 mM HEPES (pH 7.5), 150 mM NaCl, 1% Triton X-100, 0.1% SDS, 5 mM EDTA, 1 mM NaF, 200 µM sodium orthovanadate, and protease inhibitors (1 µg/ml leupeptin, 1 µg/ml aprotinin, 1 mM phenylmethylsulfonyl fluoride). The lysates were cleared from insoluble material by centrifugation at 20,000 × g for 10 min, subjected to polyacrylamide gel electrophoresis, transferred to nitrocellulose, and analyzed by Western blotting with 0.5 µg/ml primary antibodies followed by 0.3 µg/ml horseradish peroxidase-conjugated secondary antibodies and developed by ECL (Amersham Pharmacia Biotech).

Cyclic AMP Assay-- Cyclic AMP accumulation was determined as described previously (26). Briefly, cells were serum-starved and labeled with 3 µCi/ml [3H]adenine for 24 h, washed twice with serum-free DMEM, and stimulated with desired agonists for various times at 37 °C. Reactions were terminated by aspiration of medium followed by addition of ice-cold 5% trichloroacetic acid. Acid-soluble nucleotides were separated on ion-exchange columns and subjected to scintillation spectroscopy. The radioactivity of cAMP-containing fractions was normalized on the total (cAMP + ATP) radioactivity in each sample and finally expressed as -fold increase over control (zero time point).

    RESULTS
TOP
ABSTRACT
INTRODUCTION
MATERIALS AND METHODS
RESULTS
DISCUSSION
REFERENCES

ET1-induced Activation of PKA-- Fig. 1 shows a time course of PKA activation in response to ET1 (Fig. 1A) and beta 2-adrenergic receptor agonist isoproterenol (ISO) (Fig. 1B) after transient transfection of HeLa cells with ETA and beta 2-adrenergic receptor, respectively, as measured by gel retardation of the PKA substrate VASP (see "Materials and Methods"). ET1 induced a transient phosphorylation of VASP with a maximum at 5 min. In contrast, ISO-induced phosphorylation of VASP was much stronger and persisted for at least 1 h (Fig. 1B). To confirm that phosphorylation of VASP is mediated by PKA, we employed a cAMP-unresponsive dominant negative mutant of PKAr, delta R1alpha . As shown in Fig. 1C, phosphorylation of VASP, induced by ET1 and ISO, was abolished by overexpression of delta R1alpha . Confirming the specificity of delta R1alpha , it had no effect on ET1-induced MAP kinase phosphorylation (Fig. 1D) or on cGMP-mediated phosphorylation of VASP (25).


View larger version (41K):
[in this window]
[in a new window]
 
Fig. 1.   Activation of PKA by ET1 and ISO in transiently transfected HeLa cells. A and B, time course of VASP phosphorylation in response to ET1 and ISO. HeLa cells grown on 12-well plates were transfected with 100 ng of FLAG-tagged VASP cDNA, 400 ng of ETA (A), or 400 ng of beta 2-adrenergic receptor (B2AR; B) cDNAs and 1 µg of empty vector, serum-starved and stimulated with 100 nM ET1 (A) or 10 µM ISO (B) for various times. The cells were lysed, and the cell extracts were subjected to immunoblotting with anti-FLAG antibodies. C and D, effect of PKA-dominant negative mutant (delta R1alpha ) on ET1- and ISO-induced phosphorylation of VASP and MAP kinase. HeLa cells were co-transfected with cDNAs for FLAG-tagged VASP, ETA, or beta 2-adrenergic receptor as in A and B, together with 1 µg of empty vector or cDNA for delta R1alpha , as indicated. After stimulation with 100 nM ET1 or 10 µM ISO for 5 min, the cells were lysed, and the cell extracts were subjected to immunoblotting with anti-FLAG antibodies (C) or anti-phospho-MAP kinase (P-MAPK) antibodies (D). Shown are the representative blots from at least two independent experiments with similar results. P-VASP, phospho-VASP.

ET1-induced Activation of PKA Is Mediated by Degradation of Ikappa B-- Because two mechanisms of PKA activation have been described, it was important first to examine whether the effect of ET1 on PKA activity was mediated by cAMP. As shown in Fig. 2, ET1 did not stimulate cAMP production but rather reduced basal levels of cAMP in ETA-transfected HeLa cells. By contrast, ISO (positive control) increased cAMP levels by more than 8-fold in beta 2-adrenergic receptor-transfected cells (Fig. 2). This suggests that ET1-induced activation of PKA is cAMP-independent and confirms that in our cellular model, ET1 signaling is not coupled to Gs and adenylyl cyclase.


View larger version (13K):
[in this window]
[in a new window]
 
Fig. 2.   Effect of ET1 and ISO on cAMP levels in transiently transfected HeLa cells. HeLa cells grown on 12-well plates were transfected with 400 ng of ETA or 400 ng of beta 2-adrenergic receptor (B2AR) cDNAs, serum-starved and stimulated with 100 nM ET1 or 10 µM ISO for various times as indicated. The intracellular cAMP content was then measured as described under "Materials and Methods" and expressed as -fold of control. Data represent mean ± S.D. from one of two independent experiments with similar results, performed in triplicates.

We next addressed the possibility of a cAMP-independent mechanism of ET1-induced PKA activity, described previously for LPS, wherein PKA activation was mediated by proteasome-dependent degradation of Ikappa B (18). ET1 stimulated NFkappa B activity in HeLa cells by 35.8 ± 4.4-fold, as measured by kappa B-dependent expression of the luciferase gene (Fig. 3). This effect of ET1 was inhibited by the proteasome inhibitor MG-132, as well as by overexpression of the phosphorylation-deficient dominant negative mutant of Ikappa B, Ikappa Balpha m (Fig. 3). These data indicate that ET1 stimulates NFkappa B via phosphorylation and degradation of Ikappa B.


View larger version (10K):
[in this window]
[in a new window]
 
Fig. 3.   ET1-induced activation of NFkappa B. HeLa cells grown on 12-well plates were transfected with 100 ng of kappa B-driven luciferase reporter plasmid, 100 ng of pcDNA3-LacZ, 400 ng of ETA cDNA, and 400 ng of empty vector or the cDNA for Ikappa B dominant negative mutant, Ikappa Balpha m, as indicated. Quiescent cells were pretreated with or without proteasome inhibitor MG-132 (50 µM) for 1 h as indicated, followed by stimulation with 100 nM ET1 for 6 h. Luciferase activity in cell lysates was then measured, normalized on beta -galactosidase activity, and expressed as -fold activation over control (mean ± S.D. from one of three independent experiments with similar results, performed in triplicates).

Preincubation of cells with increasing concentrations of MG-132 resulted in a dose-dependent inhibition of ET1-induced PKA activity, reaching maximum at 15 µM MG-132 (Fig. 4A). By contrast, up to 50 µM MG-132 had no significant effect on ET1-induced phosphorylation of MAP kinase (Fig. 4B) or the ISO-induced VASP shift (Fig. 4F). This suggests that ET1-induced activation of PKA is mediated by proteasome-dependent protein degradation. To examine whether this PKA activation is dependent on the degradation of Ikappa B, we employed phosphorylation-deficient dominant negative mutants of Ikappa B. PKA was previously shown to bind Ikappa Balpha , as well as Ikappa Bbeta isoforms (18). Therefore, we examined the effects of Ikappa Balpha -S32A,S36A (Ikappa Balpha m) and Ikappa Bbeta -S19A,S23A (Ikappa Bbeta m) overexpression on ET1-induced PKA activity. Overexpression of increasing amounts of Ikappa Balpha m resulted in a dose-dependent inhibition of ET1-induced PKA activity (Fig. 4C) without affecting MAP kinase phosphorylation (Fig. 4D) or the ISO-induced VASP shift (Fig. 4F). By contrast, overexpression of Ikappa Bbeta m had no significant effect on ET1-induced VASP phosphorylation (Fig. 4E). Taken together, these data suggest that proteasome-dependent degradation of Ikappa Balpha mediates ET1-stimulated PKA activity in HeLa cells.


View larger version (43K):
[in this window]
[in a new window]
 
Fig. 4.   Effect of MG-132, Ikappa Balpha m, and Ikappa Bbeta m on ET1- and ISO-induced phosphorylation of VASP and MAP kinase. HeLa cells grown on 12-well plates were transfected with 100 ng of FLAG-VASP cDNA, 400 ng of ETA (A-E) or 400 ng of beta 2-adrenergic receptor (F) cDNAs, and 400 ng of empty vector or various amounts of cDNA for Ikappa Balpha m (C and D) or Ikappa Bbeta m (E), as indicated. Quiescent cells were pretreated with or without various concentrations of MG-132 for 1 h (A and B), followed by stimulation with 100 nM ET1 (A-E) or 10 µM ISO (F) for 5 min. The cells were lysed, and the cell extracts were analyzed on VASP shift (A, C, E, and F) and MAP kinase phosphorylation (B and D) by immunoblotting with anti-FLAG and anti-phospho-MAP kinase (P-MAPK) antibodies, respectively. P-VASP, phospho-VASP.

Activation of PKA by ET1 and Angiotensin II in Vascular Smooth Muscle Cells-- It was important to confirm that cAMP-independent activation of PKA by ET1 in HeLa cells was not an artifact of ETA overexpression. Therefore, we next examined the ability of ET1 to activate PKA in a primary culture of RASMC, which express endogenous levels of ETA receptors. As shown in Fig. 5A, ET1 and ISO stimulated phosphorylation of VASP in these cells with a striking similarity to their effects in the transiently transfected cellular model (compare Fig. 5A and Fig. 1). Moreover, in RASMC, PKA was also stimulated by another vasoactive peptide, angiotensin II (AII) (Fig. 5A). Importantly, ET1 and AII failed to stimulate cAMP production in RASMC, whereas ISO increased cAMP levels by more than 200-fold (Fig. 5B). This suggests that cAMP-independent activation of PKA may be a general phenomenon, common for vasoactive peptides.


View larger version (25K):
[in this window]
[in a new window]
 
Fig. 5.   Activation of PKA by ET1, AII, and ISO in vascular smooth muscle cells. Rat aortic smooth muscle cells were transfected with 200 ng of FLAG-VASP cDNA, serum-starved and stimulated with 100 nM ET1, 100 nM AII, or 10 µM ISO for various times as indicated. The cells were lysed, and the cell extracts were analyzed on VASP shift (A) or on cAMP levels (B; mean ± S.D. from one representative experiment performed in triplicate). Note the difference between A and B in stimulation time points. Shown are the representative data from at least three (A) or two (B) experiments. P-VASP, phospho-VASP.


    DISCUSSION
TOP
ABSTRACT
INTRODUCTION
MATERIALS AND METHODS
RESULTS
DISCUSSION
REFERENCES

The present study describes for the first time cAMP-independent activation of PKA by G protein-coupled receptor agonist endothelin-1 and provides the mechanism of this signaling event.

Cyclic AMP-independent Activation of PKA by Vasoactive Peptides-- Employing two independent cellular models with overexpressed or endogenous levels of ETA receptors, we provide strong evidence for the ability of ET1 to stimulate PKA activity in a cAMP-independent manner. Moreover, this may represent a general phenomenon common for vasoactive peptides, because angiotensin II elicited similar effect on PKA in RASMC. With the exception of one study, which showed a modest, cAMP-dependent activation of PKA by ET1 in pig coronary arteries (10), the stimulation of PKA by either ET1 or AII has not been reported. In our experiments, ET1 failed to stimulate cAMP production but rather reduced the basal levels of cAMP. This is in accord with other investigators having shown that ET1 either had no effect or inhibited basal or agonist-induced cAMP production, which is consistent with the coupling of ETA receptors to Gi proteins (5, 7, 13-15). However, one might still consider the possibility of compartment-specific changes in cAMP-levels in response to ET1, which have not been detected in the present study.

ET1-induced PKA Activity Is Dependent on Ikappa B Degradation-- The cAMP-independent mechanism of PKA activation, which is mediated by LPS-induced degradation of Ikappa B, has been described previously by Zhong et al. (18). However, except for bacterially derived LPS, no physiological ligand has been reported to activate PKA by this mechanism. The present work demonstrates for the first time that the physiologically relevant hormone ET1, which is central to cardiovascular, renal, and pulmonary physiology, also stimulates PKA in an Ikappa B-dependent manner (Fig. 4). This suggests that this mechanism for PKA activation is more widespread and might also be relevant to other G protein-coupled receptors.

Several important questions are still to be resolved, such as the signaling pathways, which link ETA receptors to the degradation of Ikappa B and activation of PKA, as well as the functional significance of ET1-induced PKA activation. Ikappa B degradation can be mediated by a variety of mechanisms, including protein kinase C (27, 28), mitogen-activated protein kinase (29), or Akt/protein kinase B (21). ETA receptors can activate all above-mentioned molecules (30-32), suggesting several possibilities for the signaling cascades leading to ET1-induced activation of PKA. Regarding the functional significance of ET1-induced PKA activation, stimulation of PKA by isoproterenol or forskolin was shown to inhibit agonist-induced activation of phospholipase C (33), Ca2+ mobilization (34), and Ca2+ entry (22), as well as MAP kinase cascade (7, 35), the signaling pathways commonly stimulated by G protein-coupled receptors including ETA. Moreover, it is generally accepted that activation of PKA leads to cell relaxation and regulation of cell growth (36, 37), which is opposite of vasoconstrictive and proliferative effects of ET1. This suggests that activation of PKA may serve as a regulatory mechanism in the function of ET1. Future studies will address these issues.

    ACKNOWLEDGEMENTS

We thank Dr. Masashi Yanagisawa for providing ETA receptor cDNA, Dr. Michael Uhler for providing FLAG-VASP cDNA, Dr. Stanley McKnight for providing delta R1alpha cDNA, Dr. Inder Verma for providing Ikappa Balpha m cDNA, Dr. Sergei Orlov for providing primary culture of rat aortic smooth muscle cells, and Dr. Tohru Kozasa for useful suggestions.

    FOOTNOTES

* This work was supported by National Institutes of Health Grant GM56159 and a grant from American Heart Association (to T. V. Y.).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.

Dagger To whom correspondence should be addressed: Dept. of Pharmacology (M/C 868), Medical Sciences Bldg., Rm. E-407, 835 S. Wolcott Ave., University of Illinois at Chicago, Chicago, IL 60612. Tel.: 312-355-2568; Fax: 312-996-1225; E-mail: dulin@uic.edu.

Published, JBC Papers in Press, April 30, 2001, DOI 10.1074/jbc.C100195200

    ABBREVIATIONS

The abbreviations used are: ET1, endothelin-1; Ikappa B, inhibitor of kappa B; ISO, isoproterenol; LPS, lipopolysaccharide; NFkappa B, nuclear factor kappa B; PKA, protein kinase A; RASMC, rat aortic smooth muscle cells; VASP, vasodilator-stimulated phosphoprotein; MAP, mitogen-activated protein; DMEM, Dulbecco's modified Eagle's medium; FBS, fetal bovine serum; AII, angiotensin II.

    REFERENCES
TOP
ABSTRACT
INTRODUCTION
MATERIALS AND METHODS
RESULTS
DISCUSSION
REFERENCES

1. Parris, R. J., and Webb, D. J. (1997) Vasc. Med. 2, 31-43[Medline] [Order article via Infotrieve]
2. Ortega Mateo, A., and de Artinano, A. A. (1997) Pharmacol. Res. 36, 339-351[CrossRef][Medline] [Order article via Infotrieve]
3. Adachi, M., Yang, Y. Y., Furuichi, Y., and Miyamoto, C. (1991) Biochem. Biophys. Res. Commun. 180, 1265-1272[Medline] [Order article via Infotrieve]
4. Sakamoto, A., Yanagisawa, M., Sakurai, T., Takuwa, Y., Yanagisawa, H., and Masaki, T. (1991) Biochem. Biophys. Res. Commun. 178, 656-663[Medline] [Order article via Infotrieve]
5. Vogelsang, M., Broede-Sitz, A., Schafer, E., Zerkowski, H. R., and Brodde, O. E. (1994) J. Cardiovasc. Pharmacol. 23, 344-347[Medline] [Order article via Infotrieve]
6. Takagi, Y., Ninomiya, H., Sakamoto, A., Miwa, S., and Masaki, T. (1995) J. Biol. Chem. 270, 10072-10078[Abstract/Free Full Text]
7. Wu-Wong, J. R., and Opgenorth, T. J. (1998) J. Cardiovasc. Pharmacol. 31 Suppl. 1, 185-191
8. Mao, J., Yuan, H., Xie, W., Simon, M. I., and Wu, D. (1998) J. Biol. Chem. 273, 27118-27123[Abstract/Free Full Text]
9. Aramori, I., and Nakanishi, S. (1992) J. Biol. Chem. 267, 12468-12474[Abstract/Free Full Text]
10. El-Mowafy, A. M., and White, R. E. (1998) Biochem. Biophys. Res. Commun. 251, 494-500[CrossRef][Medline] [Order article via Infotrieve]
11. Rebsamen, M. C., Church, D. J., Morabito, D., Vallotton, M. B., and Lang, U. (1997) Am. J. Physiol. 273, E922-E931[Abstract/Free Full Text]
12. Takuwa, N., Takuwa, Y., Yanagisawa, M., Yamashita, K., and Masaki, T. (1989) J. Biol. Chem. 264, 7856-7861[Abstract/Free Full Text]
13. Lin, W. W., and Chuang, D. M. (1993) Mol. Pharmacol. 44, 158-165[Abstract]
14. James, A. F., Xie, L. H., Fujitani, Y., Hayashi, S., and Horie, M. (1994) Nature 370, 297-300[Medline] [Order article via Infotrieve]
15. Zhu, Y., Yang, H. T., and Endoh, M. (1997) Am. J. Physiol. 273, H119-H127[Abstract/Free Full Text]
16. Taylor, S. S., Buechler, J. A., Yonemoto, W., Ortega Mateo, A., and de Artinano, A. A. (1990) Annu. Rev. Biochem. 59, 971-1005[CrossRef][Medline] [Order article via Infotrieve]
17. Francis, S. H., and Corbin, J. D. (1999) Crit. Rev. Clin. Lab. Sci. 36, 275-328[Medline] [Order article via Infotrieve]
18. Zhong, H., SuYang, H., Erdjument-Bromage, H., Tempst, P., and Ghosh, S. (1997) Cell 89, 413-424[Medline] [Order article via Infotrieve]
19. Baldwin, A. S., Jr. (1996) Annu. Rev. Immunol. 14, 649-683[CrossRef][Medline] [Order article via Infotrieve]
20. Ghosh, S., May, M. J., and Kopp, E. B. (1998) Annu. Rev. Immunol. 16, 225-260[CrossRef][Medline] [Order article via Infotrieve]
21. Xie, P., Browning, D. D., Hay, N., Mackman, N., and Ye, R. D. (2000) J. Biol. Chem. 275, 24907-24914[Abstract/Free Full Text]
22. Orlov, S. N., Tremblay, J., and Hamet, P. (1996) Hypertension 27, 774-780[Abstract/Free Full Text]
23. Halbrugge, M., Friedrich, C., Eigenthaler, M., Schanzenbacher, P., and Walter, U. (1990) J. Biol. Chem. 265, 3088-3093[Abstract/Free Full Text]
24. Butt, E., Abel, K., Krieger, M., Palm, D., Hoppe, V., Hoppe, J., and Walter, U. (1994) J. Biol. Chem. 269, 14509-14517[Abstract/Free Full Text]
25. Browning, D. D., McShane, M. P., Marty, C., and Ye, R. D. (2000) J. Biol. Chem. 275, 2811-2816[Abstract/Free Full Text]
26. Voyno-Yasenetskaya, T. A., Conklin, B. R., Gilbert, R. L., Hooley, R., Bourne, H. R., and Barber, D. L. (1994) J. Biol. Chem. 269, 4721-4724[Abstract/Free Full Text]
27. Coudronniere, N., Villalba, M., Englund, N., and Altman, A. (2000) Proc. Natl. Acad. Sci. U. S. A. 97, 3394-3399[Abstract/Free Full Text]
28. Sun, Z., Arendt, C. W., Ellmeier, W., Schaeffer, E. M., Sunshine, M. J., Gandhi, L., Annes, J., Petrzilka, D., Kupfer, A., Schwartzberg, P. L., and Littman, D. R. (2000) Nature 404, 402-407[CrossRef][Medline] [Order article via Infotrieve]
29. Schulze-Osthoff, K., Ferrari, D., Riehemann, K., and Wesselborg, S. (1997) Immunobiology 198, 35-49[Medline] [Order article via Infotrieve]
30. Simonson, M. S., and Dunn, M. J. (1990) FASEB J. 4, 2989-3000[Abstract]
31. Yamboliev, I. A., Hruby, A., and Gerthoffer, W. T. (1998) Pulm. Pharmacol. Ther. 11, 205-208[CrossRef][Medline] [Order article via Infotrieve]
32. Foschi, M., Chari, S., Dunn, M. J., and Sorokin, A. (1997) EMBO J. 16, 6439-6451[Abstract/Free Full Text]
33. Dodge, K. L., and Sanborn, B. M. (1998) Endocrinology 139, 2265-2271[Abstract/Free Full Text]
34. Tertyshnikova, S., and Fein, A. (1998) Proc. Natl. Acad. Sci. U. S. A. 95, 1613-1617[Abstract/Free Full Text]
35. Graves, L. M., Bornfeldt, K. E., Raines, E. W., Potts, B. C., MacDonald, S. G., Ross, R., and Krebs, E. G. (1993) Proc. Natl. Acad. Sci. U. S. A. 90, 10300-10304[Abstract]
36. Murray, K. J. (1990) Pharmacol. Ther. 47, 329-345[CrossRef][Medline] [Order article via Infotrieve]
37. Bornfeldt, K. E., and Krebs, E. G. (1999) Cell. Signal. 11, 465-477[CrossRef][Medline] [Order article via Infotrieve]


Copyright © 2001 by The American Society for Biochemistry and Molecular Biology, Inc.