Literature
首页医源资料库在线期刊动脉硬化血栓血管生物学杂志2006年第26卷第6期

Apelin Stimulates Myosin Light Chain Phosphorylation in Vascular Smooth Muscle Cells

来源:《动脉硬化血栓血管生物学杂志》
摘要:Apelin-inducedphosphorylationofMLCwasaccompaniedwithmyosinphosphatasetargetsubunitphosphorylation。【关键词】apelinAPJmyosinlightchainmyosinphosphatasetargetsubunitvasoconstrictionIntroductionApelinwasrecentlyidentifiedfrombovinestomachasanendogenousl......

点击显示 收起

【摘要】  Objective- Physiological roles of apelin and its specific receptor APJ signaling were investigated in vascular smooth muscle cells (VSMCs). The present study determined whether apelin activates myosin light chain (MLC), a major regulatory event in initiating smooth muscle contraction.

Methods and Results- To assess MLC activation, we performed Western blot and immunohistochemical studies using an antibody against the phospho-MLC. In VSMCs, apelin induces the phosphorylation of MLC in a concentration-dependent manner with a peak at 2 minutes. Pretreatment of VSMCs with pertussis toxin abolishes the apelin-induced phosphorylation of MLC. Inhibition of protein kinase C (PKC) with GF-109203X markedly attenuated the apelin-induced MLC phosphorylation. In addition, methylisobutyl amiloride, a specific inhibitor of the Na + /H + exchanger (NHE), and KB-R7943, a potent inhibitor for the reverse mode of the Na + /Ca 2+ exchanger (NCX), significantly suppressed the action of apelin. In wild-type mice, apelin phosphorylates MLC in vascular tissue, whereas it had no response in APJ-deficient mice by Western blot and immunohistochemistry. Apelin-induced phosphorylation of MLC was accompanied with myosin phosphatase target subunit phosphorylation.

Conclusions- These results provide the first evidence to our knowledge for apelin-mediated MLC phosphorylation in vitro and in vivo, which is a potential mechanism of apelin-mediated vasoconstriction.

In the vascular smooth muscles, apelin produced APJ-dependent phosphorylation of myosin light chain. This reaction was accompanied with myosin phosphatase target subunit phosphorylation. The apelin-APJ system may play a role in regulating vascular contraction.

【关键词】  apelin APJ myosin light chain myosin phosphatase target subunit vasoconstriction


Introduction


Apelin was recently identified from bovine stomach as an endogenous ligand for APJ, a putative receptor protein related to the angiotensin-type 1 receptor (AT1). 1 Despite sharing 31% amino acid sequence homology with AT1, APJ does not display specific binding for angiotensin II. 2 Apelin and APJ are distributed in various tissues including the heart, blood vessels, brain, and gastrointestinal tract, although the physiological role of apelin and APJ is not well understood. 3-7


In the vascular system, apelin and APJ are known to be expressed in endothelium and smooth muscle cells (VSMCs). Histological studies in rat show that the VSMCs of the medial layer of the aorta and pulmonary artery display intense staining for APJ receptor-like immunoreactivity. 7 In spontaneously hypertensive rats, APJ and apelin expression in both heart and aorta were markedly depressed compared with Wistar-Kyoto rats, which suggests a pathophysiological role for APJ and apelin in vascular disease. 8


The action of apelin in blood pressure regulation is controversial. Although the systemic administration of apelin produces transient hypotension in anesthetized 5,9-11 and conscious rats, 12 a potent vasoconstrictive effect of the peptide has been demonstrated in the isolated human saphenous vein. 13 These results suggest that the apelin-APJ has biphasic effects on blood vessels via the endothelium-mediated vasodilatation and VSMCs-dependent vasoconstriction. Our previous study on murine endothelium demonstrated endothelial nitric oxide synthase phosphorylation by the specific apelin-induced stimulation. 9 At the present time, however, the intracellular mechanisms of apelin have not been elucidated in VSMCs. Here, we focus on VSMCs to investigate apelin/APJ-mediated intracellular signal transduction mechanisms with particular reference to myosin light chain (MLC) phosphorylation. MLC is a major regulatory molecule for smooth muscle contraction.


Methods


Cell Culture


VSMCs from the thoracic aorta of 4-week-old male Wistar rats were prepared by the explant method and cultured in Dulbecco?s modified Eagle?s Medium as described previously. 14,15 VSMCs multiplying in a medium containing FBS rapidly lose their contractile features and become "synthetic" cells. 14 Thus, we confined our studies to early passage cells (third to the sixth passage). Subconfluent VSMCs were used in the following experiments. Cells at subconfluence were serum deprived for 24 hours and then stimulated as indicated. After the desired incubation period, cells were rinsed with ice-cold phosphate-buffered saline and then lysed and sonicated. The experiments were conducted under the guidelines for animal experiments set by the Animal Experiment Committee of the Yokohama City University School of Medicine.


Reverse-Transcriptase Polymerase Chain Reaction


Total RNA was isolated from the VSMCs by the acid guanidinium thiocyanate-phenol-chloroform extraction method and reverse-transcriptase polymerase chain reaction (RT-PCR) for APJ and GAPDH mRNA was performed; 16,17 0.3 µg of the sample RNA was reverse-transcribed using SuperScript III TM reverse transcriptase (Invitrogen, Ontario, Canada) and oligo(dT) 12 to 18 primers. APJ cDNA was amplified in PCR reactions using the following primers: sense, 5'-ATG GAA GAT GAT GGT TAC AAC TAC T-3'; antisense, 5'-CGA AAC ACG GTC CAG AGC ACC-3'. The PCR products were size fractionated on 7.5% polyacrylamide gels, stained with ethidium bromide, and then photographed. APJ-deficient mice were generated as described previously, 9 and the thoracic aorta of 10-week-old male mice were examined.


Tissue Preparation


Thoracic aortae were carefully isolated from mice after chloroform anesthesia and immediately immersed in ice-cold modified Kreb?s solution (118 mmol/L NaCl, 4.7 mmol/L KCl, 1.18 mmol/L KH 2 PO 4, 1.17 mmol/L MgSO 4, 2.5 mmol/L CaCl 2, 5.0 mmol/L HEPES, 11.1 mmol/L D-glucose, pH 7.4). Fat and connective tissues were then removed from the preparation. Endothelium was removed by gently rubbing the intima with a cotton swab to avoid the endothelium-dependent effect. Each aorta was equilibrated for 1 hour at 37°C in modified Kreb?s solution. Thereafter, the aortae were treated with or without apelin (10 -6 mol/L) for indicated periods and rinsed with ice-cold phosphate-buffered saline and then lysed and sonicated.


Western Blot


Western blot was performed to determine APJ expression and the levels of phosphorylation of MLC and myosin phosphatase target subunit (MYPT) in VSMCs and vascular tissue. Soluble proteins were fractionated on 10% SDS-polyacrylamide gels and transferred to polyvinylidene fluoride (PVDF) membranes (Millipore, Bedford, Mass). APJ protein was detected by immunoblotting with rabbit antibodies directed against mouse APJ (1:200). 18 Activation of MLC by phosphorylation on Thr 18 and/or Ser 19 was revealed by immunoblotting with rabbit polyclonal antibodies against the phospho-MLC (1:1000; Santa Cruz Biotechnology, Santa Cruz, Calif), whereas total MLC protein was detected using goat polyclonal antibodies against total MLC (1:1000; Santa Cruz Biotechnology). 19,20 Activation of MYPT by phosphorylation on Thr 696 was revealed by immunoblotting with rabbit polyclonal antibodies against the phospho-MYPT (1:200; Santa Cruz Biotechnology), whereas total MYPT protein was detected using rabbit polyclonal antibodies against total MYPT (1:200; Santa Cruz Biotechnology). Immunoreactive bands were visualized with specific antibodies using ECL plus reagent (Amersham Biosciences, Piscataway, NJ), and the images were analyzed by densitometry using Scion Image Software (Scion Corporation, Frederick, Md). 21,22 Pertussis toxin (PTX) (Seikagaku Co, Tokyo, Japan), GF109203X (Tocris, Ellisville, Mo), U-73122 (Calbiochem, Darmstadt, Germany), methylisobutyl amiloride (MIA) (Sigma-Aldrich, St Louis, Mo), and KB-R7943 (Tocris) were added to the culture 24 hours, 1 hour, 1 hour, 5 minutes, and 5 minutes before [Pyr 1 ]-apelin-13 (Peptide Institute, Osaka, Japan) stimulation, respectively.


Immunohistochemistry


Immunohistochemistry for formalin-fixed aorta treated with or without apelin was performed as follows. Sections of 3 µm were cut and mounted on glass slides. The endogenous peroxidase was blocked with 3% H 2 O 2, and nonspecific binding sites were blocked with TBS containing 5% normal goat serum. Slides were then incubated with rabbit polyclonal antibody against the phospho-MLC (1:200; Santa Cruz Biotechnology) for 1 hour at room temperature. For negative controls, the primary antibody was replaced with control rabbit IgG (Santa Cruz Biotechnology). Staining was visualized with the avidin-biotin immunoperoxidase reaction using diaminobenzidine (LSAB2 Kit; DAKO, Glostrup, Denmark), and the images were analyzed by staining densitometry using Scion Image Software (Scion Corporation) as described previously. 21,22


Statistical Analysis


Data are expressed as mean±SEM. Statistical significance was determined by ANOVA with P <0.05 being deemed statistically significant.


Results


Expression of APJ in VSMCs


Gene expression of APJ in VSMCs was analyzed by RT-PCR. As shown in Figure 1 A, the APJ mRNA was amplified using RNA prepared from cultured rat VSMCs and from the aorta of wild-type mice. No amplified product was detected using RNA prepared from the aorta of APJ-deficient mice. The APJ antibody detected a band at 50 kDa in isolated thoracic aorta of wild-type mice ( Figure 1 B). There was no detectable band in the aorta of APJ-deficient mice. This is consistent with a previous report showing APJ receptor expression in HEK-293 cells stably expressing the APJ. 6


Figure 1. APJ is expressed in VSMCs and isolated aorta. APJ mRNA expression was assessed by RT-PCR (A). Total RNA was isolated, and RT-PCR was performed using primers specific for APJ. The PCR products were size-fractionated on a polyacrylamide gel that was stained with ethidium bromide. The 158-bp PCR product was amplified from VSMCs RNA. No product was detected using RNA isolated from the aorta of APJ-deficient mice. APJ protein expression was also assessed by Western blot (B). Total protein was isolated and the Western blot was probed using antibody directed against mouse APJ (1:200) raised in rabbit.


Effect of Apelin on MLC Phosphorylation in Cultured VSMCs


Activation of MLC is regulated by phosphorylation. Ser 19 of MLC has been shown to be the primary residue phosphorylated by MLC kinase, with Thr 18 as a secondary site. Western blot analysis showed apelin to induce MLC phosphorylation at Thr 18 and/or Ser 19. The phosphorylation reaction reached a maximal level 2 minutes after the addition of apelin (10 -7 mol/L) ( Figure 2 A). Furthermore, the reaction proceeded in a concentration-dependent (0 to 10 -5 mol/L) manner ( Figure 2 B).


Figure 2. Time course of phosphorylation of MLC by apelin in VSMCs is shown (A). VSMCs were starved for 24 hours, stimulated with 10 -7 mol/L apelin for the indicated periods of time, and then lysed and analyzed by SDS-PAGE and immunoblotting for Thr 18/Ser 19 phosphorylated MLC (A, upper panel) or total MLC protein (A, lower panel) using specific antisera. Apelin promotes a concentration-dependent phosphorylation of MLC (B). VSMCs were stimulated with varying concentrations of apelin for 2 minutes and lysates were immunoblotted as described previously. The intensity of each band on the blot was quantified by densitometric scanning, and the activities of MLC are shown as fold increases of the average from 3 independent experiments by comparison with nonstimulated controls (1.0). * P <0.05 vs nonstimulated controls.


Inhibitors of G Protein, Protein Kinase C, or Ion Exchangers


In the following experiments, the effects of drugs were evaluated 2 minutes after the stimulation of apelin (10 -7 mol/L). To examine intracellular signal transduction mechanisms, VSMCs were pretreated with inhibitors for G protein, PKC, and ion exchangers.


As shown in Figure 3, PTX (100 ng/mL), which uncouples Gi/Go proteins from their receptors by ADP-ribosylation, abolished the apelin-induced phosphorylation of MLC. GF109203X (10 -5 mol/L), a specific inhibitor of PKC, also abolished the apelin-induced phosphorylation of MLC. U-73122 (10 -5 mol/L), a potent inhibitor of phospholipase C (PLC), had no influence on MLC phosphorylation, although it abolished the angiotensin II-induced phosphorylation of MLC (data not shown). In the absence of apelin, the addition of PTX, GF109203X or U-73122 did not affect MLC phosphorylation (data not shown).


Figure 3. Phosphorylation of MLC by apelin involves a PTX-sensitive protein and PKC. VSMCs were treated with PTX (100 ng/mL) for 24 hours (A), GF109203X (10 -5 mol/L) for 1 hour (B) or U-73122 (10 -5 mol/L) for 1 hour (C), followed by stimulation with apelin (10 -7 mol/L) for 2 minutes. MLC activity was measured as described in the legend to Figure 2. The intensity of each band on the blot was quantified by densitometric scanning, and the activities of MLC are shown as fold increases of the average from three independent experiments compared with unstimulated controls (1.0). * P <0.05 vs nonstimulated controls, ** P <0.05 vs apelin-stimulated groups.


MIA (10 -5 mol/L), an inhibitor of the Na + -H + exchanger (NHE), and KB-R7943 (10 -5 mol/L), an inhibitor of the Na + -Ca 2+ exchanger (NCX), also significantly attenuated the overall apelin-induced MLC phosphorylation (supplemental Figure I, please see http://atvb.ahajournals.org.). However, the effects of these inhibitors were not complete; densitometric analysis of 3 separate experiments revealed that the phosphorylation reaction was reduced to 73% and 81% of the control by MIA and KB-R7943, respectively. In the absence of apelin, MIA and KB-R7943 did not affect MLC phosphorylation (data not shown).


Effect of Apelin on MLC Phosphorylation in Isolated Thoracic Aorta


Thoracic aortae were isolated and treated with or without apelin (10 -6 mol/L). Western blot showed apelin to induce MLC phosphorylation. The phosphorylation reaction reached a maximal level 2 minutes after the addition of apelin (10 -6 mol/L) ( Figure 4 A). The MLC phosphorylation was accompanied with MYPT phosphorylation ( Figure 4 B). Immunohistochemistry showed MLC phosphorylation in the medial layer of aorta from wild-type mice ( Figure 5 A). Positive immunostaining for phospho-MLC is highly localized in the medial layer of the aorta treated with apelin ( Figure 5 B). The staining area of phospho-MLC immunoreactivity in APJ-deficient mice treated with apelin was similar to those found in none-treated wild-type mice ( Figure 5 C). Figure 5 D shows phospho-MLC expression in the wild-type mice treated with angiotensin II (10 -6 mol/L) as a positive control. In the control experiments, where the primary antibody was replaced with normal rabbit IgG, the samples were negative for phospho-MLC ( Figure 5 E). Removal of the endothelium was verified by HE staining ( Figure 5 F). These observations were quantitatively evaluated ( Figure 5 G). In wild-type mice, we detected 1.9 times more phospho-MLC staining area in the aorta treated with apelin than with nontreated aorta. We detected no differences in phospho-MLC staining area between nontreated wild-type mice aorta and APJ-deficient mice aorta treated with apelin. These results indicate apelin induces significant phosphorylation of MYPT and MLC in aorta via the APJ receptor.


Figure 4. Time course of phosphorylation of MLC by apelin in aorta is shown (A). Aortae were stimulated with 10 -6 mol/L apelin for the indicated periods of time and then lysed and analyzed by SDS-PAGE and immunoblotting for Thr 18/Ser 19 phosphorylated MLC (A, upper panel), total MLC protein (A, lower panel), Thr 696 phosphorylated MYPT (B, upper panel), and total MYPT (B, lower panel) using specific antisera. The intensity of each band on the blot was quantified by densitometric scanning, and the activities of MLC or MYPT are shown as fold increases of the average from 3 independent experiments by comparison with nonstimulated controls (1.0). * P <0.05 vs nonstimulated controls.


Figure 5. Representative photomicrographs showing phospho-MLC immunoreactivity in the medial layer of aorta from wild-type (A, B, D) and APJ-deficient (C) mice treated with apelin (10 -6 mol/L) (B, C), without apelin (A) or with angiotensin II (10 -6 mol/L) (D). Positive immunostaining for phospho-MLC is highly localized in the medial layer of the aorta treated with apelin (B). Staining area for phospho-MLC in the medial layer of aorta significantly increased in response to the apelin treatment. No such response was observed in the aorta from APJ-defient mice (G). n=6 in each group. * P <0.05 compared with the wild-type mice without apelin. P <0.05 compared with the wild-type mice with apelin.


Discussion


In the present study we have demonstrated that APJ is expressed in aortic VSMCs at the mRNA and protein level. This is consistent with a previous study, which showed the autoradiographical localization of apelin binding in the medial layer of the human coronary artery and saphenous vein 13 and immunohistochemical staining for APJ in human and rat blood vessels. 7


Primary mechanisms for the contraction of smooth muscle are the phosphorylation of MLC by MLC kinase and inhibition of MLC phosphatase. The transient increase in MLC phosphorylation, a biochemical index of vascular contraction, can be stimulated in vascular smooth muscle preparations by a variety of vasoactive hormones and neurotransmitters, including angiotensin II, histamine, -adrenergic agonists, and neuropeptide Y. 23 Apelin is derived from a 77-amino-acid precursor, and processed to several isoforms by modifying the amino terminus. 3,4 The pyroglutamylated form of apelin-13, [Pyr 1 ]-apelin-13, has been reported to be produced in vivo in bovine colostrums and have activity at the receptor. 3 In our experiment, apelin induced significant phosphorylation of MLC in cultured rat VSMCs in a dose-dependent fashion. The in vitro response to apelin stimulation is the same in the VSMCs of the intact arterial media. Apelin enhanced the phospho-MLC immunoreactivities in freshly isolated thoracic aortae of wild-type mice, whereas the same dose of the peptide had no effect on aortae of APJ-deficient mice. Immunohistochemical staining for phospho-MLC using wild-type mice and APJ-deficient mice strengthened that apelin induced phosphorylation of MLC. Apelin-induced phosphorylation of MLC was accompanied with MYPT phosphorylation, which inhibits MLC phosphatase activity and induce phosphorylation of MLC. These results suggest that apelin functions as a regulator for vasoconstriction via APJ receptors.


It has been shown that apelin/APJ signaling is coupled to PTX-sensitive G proteins (Gi or Go protein) in cultured cells. 3,18,24 In the present study, we have demonstrated apelin-stimulated MLC phosphorylation in VSMCs in a PTX-sensitive manner. This result suggests that apelin-APJ signaling mediates Gi or Go protein to phosphorylate MLC in VSMCs. The mode of action of apelin may be like other Gi or Go protein-linked vasoconstrictors such as sphingolipids, 25 noradrenaline, 26,27 and neuropeptide Y. 28


It has been demonstrated that PKC enhances the phosphorylation of MLC at multiple sites in smooth muscle 29 and promotes contraction of the aorta. 30 In our experiments, apelin-induced MLC phosphorylation was markedly attenuated by GF109203X, a specific inhibitor of PKC, suggesting that PKC activates apelin-induced MLC phosphorylation. PKC can activate various target proteins in VSMCs. 31,32 Among them, NHE and NCX are known to play important roles in tonic contraction of arterial smooth muscle cells, such that activated NHE mediates the elevation of the cytosolic concentration of sodium, leading to an influx of calcium through NCX. 33 We showed that MIA and KB-R7943 markedly reduced the MLC phosphorylation stimulated by apelin in VSMCs, suggesting the involvement of NHE and NCX in apelin-induced MLC phosphorylation. The apelin-induced increase in developed tension of isolated rat heart was partly reduced by PTX and markedly attenuated by U-73122, a potent inhibitor of phospholipase C. 34 In our experiments, PTX significantly lowered apelin-induced MLC phosphorylation in VSMCs. However, U-73122 failed to inhibit the apelin-induced phosphorylation of MLC. The coupling of G protein with APJ appears to be different in VSMCs and heart.


This is the first report to demonstrate the potential mechanism of apelin-induced vasoconstriction. Our findings are consistent with a previous report showing that apelin potently contracts the human saphenous vein. 13 However, we and others reported that blood pressure is transiently lowered by the systemic administration of apelin 5,10,12 via APJ, 9 which involves the activation of the nitric oxide/L-arginine system. 11 Because APJ-mediated apelin stimulation phosphorylates endothelial nitric oxide synthase in cultured endothelial cells, 9 the apelin-induced vasodilatation may depend on the endothelial cells. Both VSMCs and endothelial cells express APJ, suggesting a biphasic action. From a pathophysiological point of view, it might be speculated that apelin functions as a vasopressor in damaged vasculature (eg, atherosclerosis). A recent report showed that insulin upregulates apelin mRNA expression in adipocytes and both plasma apelin and insulin levels were significantly higher in obese patients. 35 Additional studies will determine whether apelin is involved in the obesity-associated vascular damage.


In conclusion, we have demonstrated that APJ is expressed in VSMCs, and that apelin induces the phosphorylation of MLC in vitro and in vivo. The apelin-induced MLC phosphorylation may involve PTX-sensitive G proteins, and activation of PKC, NHE, NCX, and MYPT. This is the first report for the potential mechanism of vasoconstriction by APJ in VSMCs.


Acknowledgments


This study was supported by grants from the 21st Century COE Program, from the Japan Society for the Promotion of Science Grant (JSPS), a grant-in-aid for Scientific Research from the JSPS (2002, 13670735) (2004, 16590704), a grant-in-aid for Young Scientists from the Ministry of Education, Culture, Sports, Science and Technology of Japan (2004, 16790425), and from the Yokohama Foundation for Advancement of Medical Science. We acknowledge Dr Akihide Ryo, Department of Molecular Pathology, Emi Maeda, and Hiroko Morinaga, Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine and School of Medicine for scientific discussions, encouragement, and technical assistance.

【参考文献】
  Tatemoto K, Hosoya M, Habata Y, Fujii R, Kakegawa T, Zou MX, Kawamata Y, Fukusumi S, Hinuma S, Kitada C, Kurokawa T, Onda H, Fujino M. Isolation and characterization of a novel endogenous peptide ligand for the human APJ receptor. Biochem Biophys Res Commun. 1998; 251: 471-476.

O?Dowd BF, Heiber M, Chan A, Heng HH, Tsui LC, Kennedy JL, Shi X, Petronis A, George SR, Nguyen T. A human gene that shows identity with the gene encoding the angiotensin receptor is located on chromosome 11. Gene. 1993; 136: 355-360.

Hosoya M, Kawamata Y, Fukusumi S, Fujii R, Habata Y, Hinuma S, Kitada C, Honda S, Kurokawa T, Onda H, Nishimura O, Fujino M. Molecular and functional characteristics of APJ. Tissue distribution of mRNA and interaction with the endogenous ligand apelin. J Biol Chem. 2000; 275: 21061-21067.

Kawamata Y, Habata Y, Fukusumi S, Hosoya M, Fujii R, Hinuma S, Nishizawa N, Kitada C, Onda H, Nishimura O, Fujino M. Molecular properties of apelin: tissue distribution and receptor binding. Biochim Biophys Acta. 2001; 1538: 162-171.

Reaux A, De Mota N, Skultetyova I, Lenkei Z, El Messari S, Gallatz K, Corvol P, Palkovits M, Llorens-Cortes C. Physiological role of a novel neuropeptide, apelin, and its receptor in the rat brain. J Neurochem. 2001; 77: 1085-1096.

Medhurst AD, Jennings CA, Robbins MJ, Davis RP, Ellis C, Winborn KY, Lawrie KW, Hervieu G, Riley G, Bolaky JE, Herrity NC, Murdock P, Darker JG. Pharmacological and immunohistochemical characterization of the APJ receptor and its endogenous ligand apelin. J Neurochem. 2003; 84: 1162-1172.

Kleinz MJ, Skepper JN, Davenport AP. Immunocytochemical localisation of the apelin receptor, APJ, to human cardiomyocytes, vascular smooth muscle and endothelial cells. Regul Pept. 2005; 126: 233-240.

Zhong JC, Huang DY, Liu GF, Jin HY, Yang YM, Li YF, Song XH, Du K. Effects of all-trans retinoic acid on orphan receptor APJ signaling in spontaneously hypertensive rats. Cardiovasc Res. 2005; 65: 743-750.

Ishida J, Hashimoto T, Hashimoto Y, Nishiwaki S, Iguchi T, Harada S, Sugaya T, Matsuzaki H, Yamamoto R, Shiota N, Okunishi H, Kihara M, Umemura S, Sugiyama F, Yagami K, Kasuya Y, Mochizuki N, Fukamizu A. Regulatory roles for APJ, a seven-transmembrane receptor related to angiotensin-type 1 receptor in blood pressure in vivo. J Biol Chem. 2004; 279: 26274-26279.

Lee DK, Cheng R, Nguyen T, Fan T, Kariyawasam AP, Liu Y, Osmond DH, George SR, O?Dowd BF. Characterization of apelin, the ligand for the APJ receptor. J Neurochem. 2000; 74: 34-41.

Tatemoto K, Takayama K, Zou MX, Kumaki I, Zhang W, Kumano K, Fujimiya M. The novel peptide apelin lowers blood pressure via a nitric oxide-dependent mechanism. Regul Pept. 2001; 99: 87-92.

Cheng X, Cheng XS, Pang CC. Venous dilator effect of apelin, an endogenous peptide ligand for the orphan APJ receptor, in conscious rats. Eur J Pharmacol. 2003; 470: 171-175.

Katugampola SD, Maguire JJ, Matthewson SR, Davenport AP. [ 125 I]-(Pyr 1 )Apelin-13 is a novel radioligand for localizing the APJ orphan receptor in human and rat tissues with evidence for a vasoconstrictor role in man. Br J Pharmacol. 2001; 132: 1255-1260.

Hashimoto T, Kihara M, Sato K, Imai N, Tanaka Y, Sakai M, Tamura K, Hirawa N, Toya Y, Kitamura H, Umemura S. Heparin recovers AT1 receptor and its intracellular signal transduction in cultured vascular smooth muscle cells. FEBS lett. 2005; 579: 281-284.

Hashimoto T, Kihara M, Yokoyama K, Fujita T, Kobayashi S, Matsushita K, Tamura K, Hirawa N, Toya Y, Umemura S. Lipoxygenase products regulate nitric oxide and inducible nitric oxide synthase production in interleukin-1beta stimulated vascular smooth muscle cells. Hypertens Res. 2003; 26: 177-184.

Hashimoto T, Kihara M, Sato K, Matsushita K, Tanimoto K, Toya Y, Fukamizu A, Umemura S. Expression of Cyclooxygenase-2 in the Juxtaglomerular Apparatus of Angiotensinogen Gene-Knockout Mice. Nephron Physiol. 2006; 102: 1-8.

Kihara M, Umemura S, Kadota T, Yabana M, Tamura K, Nyuui N, Ogawa N, Murakami K, Fukamizu A, Ishii M. The neuronal isoform of constitutive nitric oxide synthase is up-regulated in the macula densa of angiotensinogen gene-knockout mice. Lab Invest. 1997; 76: 285-294.

Hashimoto Y, Ishida J, Yamamoto R, Fujiwara K, Asada S, Kasuya Y, Mochizuki N, Fukamizu A. G protein-coupled APJ receptor signaling induces focal adhesion formation and cell motility. Int J Mol Med. 2005; 16: 787-792.

Yuan SY, Wu MH, Ustinova EE, Guo M, Tinsley JH, De Lanerolle P, Xu W. Myosin light chain phosphorylation in neutrophil-stimulated coronary microvascular leakage. Circ Res. 2002; 90: 1214-1221.

Edens HA, Levi BP, Jaye DL, Walsh S, Reaves TA, Turner JR, Nusrat A, Parkos CA. Neutrophil transepithelial migration: evidence for sequential, contact-dependent signaling events and enhanced paracellular permeability independent of transjunctional migration. J Immunol. 2002; 169: 476-486.

Sato K, Kihara M, Hashimoto T, Matsushita K, Koide Y, Tamura K, Hirawa N, Toya Y, Fukamizu A, Umemura S. Alterations in renal endothelial nitric oxide synthase expression by salt diet in angiotensin type-1a receptor gene knockout mice. J Am Soc Nephrol. 2004; 15: 1756-1763.

Kihara M, Sato K, Hashimoto T, Imai N, Toya Y, Umemura S. Expression of endothelial nitric oxide synthase is suppressed in the renal vasculature of angiotensinogen-gene knockout mice. Cell Tissue Res. 2005; 28: 1-8.

Hathaway DR, March KL, Lash JA, Adam LP, Wilensky RL. Vascular smooth muscle. A review of the molecular basis of contractility. Circulation. 1991; 83: 382-390.

Masri B, Lahlou H, Mazarguil H, Knibiehler B, Audigier Y. Apelin (65-77) activates extracellular signal-regulated kinases via a PTX-sensitive G protein. Biochem Biophys Res Commun. 2002; 290: 539-545.

Zhou H, Murthy KS. Distinctive G protein-dependent signaling in smooth muscle by sphingosine 1-phosphate receptors S1P1 and S1P2. Am J Physiol Cell Physiol. 2004; 286: C1130-C1138.

Johnson RM, Lincoln TM. Effects of nitroprusside, glyceryl trinitrate, and 8-bromo cyclic GMP on phosphorylase a formation and myosin light chain phosphorylation in rat aorta. Mol Pharmacol. 1985; 27: 333-342.

Abebe W, Edwards JD, Agrawal DK. G-proteins in rat blood vessels-II. Assessment of functional involvement. Gen Pharmacol. 1995; 26: 75-83.

Lobaugh LA, Blackshear PJ. Neuropeptide Y stimulation of myosin light chain phosphorylation in cultured aortic smooth muscle cells. J Biol Chem. 1990; 265: 18393-18399.

Colburn JC, Michnoff CH, Hsu LC, Slaughter CA, Kamm KE, Stull JT. Sites phosphorylated in myosin light chain in contracting smooth muscle. J Biol Chem. 1988; 263: 19166-19173.

Rohra DK, Yamakuni T, Ito S, Saito SY, Ohizumi Y. Evidence for the involvement of protein kinase C in acidic pH-induced contraction in spontaneously hypertensive rat aorta. Pharmacology. 2004; 71: 10-16.

Soleimani M, Singh G, Dominguez JH, Howard RL. Long-term high osmolality activates Na(+)-H+ exchange and protein kinase C in aortic smooth muscle cells. Circ Res. 1995; 76: 530-535.

Williams B, Howard RL. Glucose-induced changes in Na+/H+ antiport activity and gene expression in cultured vascular smooth muscle cells. Role of protein kinase C. J Clin Invest. 1994; 93: 2623-2631.

Blaustein MP, Lederer WJ. Sodium/calcium exchange: its physiological implications. Physiol Rev. 1999; 79: 763-854.

Szokodi I, Tavi P, Foldes G, Voutilainen-Myllyla S, Ilves M, Tokola H, Pikkarainen S, Piuhola J, Rysa J, Toth M, Ruskoaho H. Apelin, the novel endogenous ligand of the orphan receptor APJ, regulates cardiac contractility. Circ Res. 2002; 91: 434-440.

Boucher J, Masri B, Daviaud D, Gesta S, Guigne C, Mazzucotelli A, Castan-Laurell I, Tack I, Knibiehler B, Carpene C, Audigier Y, Saulnier-Blache JS, Valet P. Apelin, a newly identified adipokine up-regulated by insulin and obesity. Endocrinology. 2005; 146: 1764-1771.


作者单位:Department of Pathology, Division of Cellular Pathobiology, and Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine and School of Medicine, Yokohama, and the Center for Tsukuba Advanced Research Alliance, Graduate School of Life and Environmen

作者: Tatsuo Hashimoto; Minoru Kihara; Junji Ishida; Noz
医学百科App—中西医基础知识学习工具
  • 相关内容
  • 近期更新
  • 热文榜
  • 医学百科App—健康测试工具