• 沒有找到結果。

Mechanisms involved in the antiplatelet activity of tetramethylpyrazine in human platelets.

N/A
N/A
Protected

Academic year: 2021

Share "Mechanisms involved in the antiplatelet activity of tetramethylpyrazine in human platelets."

Copied!
9
0
0

加載中.... (立即查看全文)

全文

(1)

Mechanisms involved in the antiplatelet activity

of magnesium in human platelets

J o e n - R o n g S h e u ,1,2G e o r g e H s i a o ,2M i n g - Y i S h e n ,1T s o r n g - H a r n F o n g ,1Y i - W i n C h e n ,1 C h i e n - H u a n g L i n1a n d D u e n - S u e y C h o u2 1Graduate Institute of Medical Sciences and2Department of Pharmacology, Taipei Medical University, Taipei, Taiwan

Received 22 April 2002; accepted for publication 15 July 2002

Summary. In this study, magnesium sulphate dose-dependently (0Æ6–3Æ0 mmol/l) inhibited platelet aggregation in human platelets stimulated by agonists. Furthermore, magnesium sulphate (3Æ0 mmol/l) markedly interfered with the binding of fluorescein isothiocanate-triflavin to the glycoprotein (GP)IIb/IIIa complex in platelets stimulated by collagen. Magnesium sulphate (1Æ5 and 3Æ0 mmol/l) also inhibited phosphoinositide breakdown and intracellular Ca+2 mobilization in human platelets stimulated by colla-gen. Magnesium sulphate (3Æ0 mmol/l) significantly inhib-ited thromboxane A2 formation stimulated by collagen

in platelets. Moreover, magnesium sulphate (1Æ5 and 3Æ0 mmol/l) obviously increased the fluorescence of platelet membranes tagged with diphenylhexatriene. In addition, magnesium sulphate (1Æ5 and 3Æ0 mmol/l) increased the formation of cyclic adenosine monophosphate (AMP) in

platelets. Phosphorylation of a protein of Mr 47 000 (P47) was markedly inhibited by magnesium sulphate (1Æ5 mmol/l). In conclusion, the antiplatelet activity of magnesium sulphate may involve the following two path-ways. (1) Magnesium sulphate may initially induce mem-brane fluidity changes with resulting interference of fibrinogen binding to the GPIIb/IIIa complex, followed by inhibition of phosphoinositide breakdown and thromboxane A2 formation, thereby leading to inhibition of both

intra-cellular Ca2+mobilization and phosphorylation of P47. (2) Magnesium sulphate might also trigger the formation of cyclic AM, ultimately resulting in inhibition of the phos-phorylation of P47 and intracellular Ca+2mobilization. Keywords: magnesium sulphate, platelet aggregation, GPIIb/IIIa complex, phospholipase C, protein kinase C.

Intravascular thrombosis is one of the generators of a wide variety of cardiovascular diseases. Initiation of an intralu-minal thrombosis is believed to involve platelet adherence and aggregation. In normal circulation, platelets cannot aggregate by themselves. However, when a blood vessel is damaged, platelets adhere to the disrupted surface, and the adherent platelets release some biologically active constit-uents and aggregate (Sheu et al, 1994). Thus, platelet aggregation may play a crucial role in the atherothrombotic process. Indeed, antiplatelet agents (e.g. ticlopidine and aspirin) have been shown to reduce the incidence of stroke in high-risk patients (Hass et al, 1989).

Magnesium is an important intracellular cation and an obligatory cofactor of many enzymes in the human body. Magnesium deficiency and its association with platelet hyper-reactivity have been well recognized in a variety of

diseases, including acute myocardial infarction (Rasmussen et al, 1988), pre-eclampsia (Lucas et al, 1995) and diabetes mellitus (Nadler et al, 1992). Magnesium has been shown to reduce platelet aggregation both in vitro and ex vivo (Hwang et al, 1992; Ravn et al, 1996a,b). Furthermore, in vitro studies have shown reduced platelet release of b-thrombo-globulin and thromboxane B2with increasing magnesium

concentrations (0Æ5–8Æ0 mmol/l) (Hwang et al, 1992; Ravn et al, 1996a,b). In addition, magnesium has also been shown to reduce thrombin-stimulated Ca+2influx in plate-lets (Hwang et al, 1992). Shechter et al (2000) reported that oral magnesium therapy significantly improved the endot-helial function of coronary artery diseases. Apart from the direct antiplatelet effect, an antithrombotic effect can also be derived from nitric oxide and prostacyclin, as magnesium has been shown to stimulate the release of these vasodilating and anti-aggregatory substances from the endothelium (Watson et al, 1986; Nadler et al, 1987).

On the other hand, studies of the mechanisms of magnesium in antiplatelet aggregation have rarely been compared with results from other antiplatelet drugs (such as aspirin and ticlopidine). We therefore systematically

Correspondence: Dr Joen-Rong Sheu, Graduate Institute of Medical Sciences and Department of Pharmacology, Taipei Medical Uni-versity, no 250, Wu-Hsing Street, Taipei 110, Taiwan. E-mail: sheujr@tmu.edu.tw

(2)

examined the influence of magnesium on washed human platelets and utilized the findings to characterize the mechanisms involved.

MATERIALS AND METHODS

Materials. Collagen (Type I, bovine achilles tendon), adenosine 5¢-diphosphate (ADP), magnesium sulphate, arachidonic acid, EDTA, luciferin–luciferase, Dowex-1 (100–200 mesh; X8, chloride form), prostaglandin E1, trichloroacetic acid, EGTA, bovine serum albumin, acryla-mide, sodium pyruvate, b-NADH, diphenylhexatriene (DPH), apyrase, heparin, thrombin and myoinositol were purchased from Sigma Chemicals (St Louis, MO, USA). Fura 2-AM and fluorescein isothiocyanate (FITC) were purchased from Molecular Probe (Eugene, OR, USA). Trimeresurus flavoviridis venom was purchased from Latoxan (Rosans, France). Myo-2-[3H] inositol was purchased from Amer-sham (Buckinghamshire, HP, UK). Thromboxane B2, cyclic

adenosine monophosphate (AMP) and cyclic guanosine monophosphate (GMP) enzyme immunoassay (EIA) kits were purchased from Cayman (Ann Arbor, MI, USA).

Preparation of human platelet suspensions. Human platelet suspensions from human volunteers who had given informed consent, were prepared as previously described (Huang et al, 1991a). Briefly, blood was collected from healthy human volunteers, who had taken no medicine during the preceding 2 weeks, and was mixed with acid/ citrate/glucose (9:1, v/v). After centrifugation at 120 g for 10 min at room temperature, the supernatant (platelet-rich plasma; PRP) was supplemented with prostaglandin E1 (PGE1) (0Æ5 lmol/l) and heparin (6Æ4 IU/ml), then incubated

for 10 min at 37C and centrifuged at 500 g for 10 min. The washed platelets were finally suspended in Tyrode’s solution, containing bovine serum albumin (BSA) (3Æ5 mg/ml), and adjusted to about 4Æ5· 108

platelets/ml. The final concen-tration of Ca2+in Tyrode’s solution was 1 mmol/l.

Platelet aggregation. The turbidimetric method used a Lumi-Aggregometer (Payton, Canada) as described previ-ously (Born & Cross, 1963). Briefly, platelet suspensions (0Æ4 ml) were prewarmed to 37C for 2 min and then magnesium sulphate was added for 3 min before the addition of platelet-aggregation inducers. The reaction was allowed to proceed for at least 6 min and the extent of aggregation was expressed as a percentage of the control (in the absence of magnesium sulphate). The degree of aggre-gation was expressed in light-transmission units. While measuring ATP release, 20ll of a luciferin/luciferase mixture was added 1 min before the addition of agonists, and ATP release was compared with that of the control.

Analysis of the platelet surface glycoprotein (GP) IIb/IIIa complex by flow cytometry. Triflavin, a specific fibrinogen receptor (GPIIb/IIIa complex) antagonist, was prepared as previously described (Sheu et al, 1992). FITC-conjugated triflavin was also prepared as previously described (Sheu et al, 1996) and the final concentration was adjusted to 1 mg/ml. Human platelet suspensions were prepared as described above. Aliquots of platelet suspensions (4Æ5· 108

/ml) were preincubated with magnesium sulphate (3Æ0 mmol/l) for

3 min, followed by the addition of 2ll of FITC-triflavin. The suspensions were then incubated for another 5 min and the volume was adjusted to 1 ml/tube with Tyrode’s solution. The suspensions were then assayed for fluorescein-labelled platelets with a flow cytometer (FACScan System; Becton Dickinson, San Jose, CA, USA). Data were collected from 50 000 platelets per experimental group. All experiments were repeated at least five times to ensure reproducibility.

Labelling of membrane phospholipids and measurement of the production of [3H]-inositol phosphates. The method was

carried out as previously described (Huang et al, 1991b). Briefly, citrated human PRP was centrifuged and the pellets were suspended in Tyrode’s solution containing [3 H]-inositol (2Æ775 MBq/ml). Platelets were incubated for 2 h followed by centrifugation and finally resuspended in Ca2+ -free Tyrode’s solution (1· 109

platelets/ml). Magnesium sulphate was preincubated with 1 ml of loaded platelets at room temperature for 3 min and collagen (1lg/ml) was then added to trigger aggregation. The reaction was stopped after 6 min and the samples were centrifuged at 1000 g for 4 min. The inositol phosphates of the supernatants were separated in a Dowex-1 anion exchange column. Only [3 H]-inositol monophosphate (IP) was measured as an index of total inositol phosphate formation.

Measurement of platelet [Ca2+]i mobilization by Fura 2-AM fluorescence. Citrated whole blood was centrifuged at 120 g for 10 min. The supernatant was protected from light and incubated with Fura 2-AM (5lmol/l) at 37C for 1 h. Human platelet suspensions were then prepared as des-cribed above. Finally, the external Ca2+concentration of the platelet suspensions was adjusted to 1 mmol/l. The rise in [Ca2+]i was measured using a fluorescence spectrophoto-meter (CAF 110; Jasco, Tokyo, Japan) with excitation wavelengths of 340 and 380 nm, and an emission wave-length of 500 nm. The [Ca2+]i was calculated from the

fluorescence measured using 224 nmol/l as the Ca2+–Fura 2 dissociation constant (Grynkiewicz et al, 1985).

Measurement of membrane fluidity by fluorescent probe. The intensity of fluorescence in human platelets was measured as described previously (Kitagawa et al, 1984). Briefly, platelets (4Æ5· 108

/ml) were mixed with 9 volumes of sodium/potassium-Tris medium. A solution of DPH in dimethyl formamide was added to the suspension at a final concentration of 0Æ5 lmol/l. Platelets were preincubated with various concentrations of magnesium sulphate (1Æ5 and 3Æ0 mmol/l) for 3 min followed by the addition of DPH at 37C for another 6 min. The relative fluorescence intensity of platelets was measured in a fluorescence spectrophotometer (Hitachi F4500, Tokyo, Japan) at 37C. Measurement of thromboxane B2 formation. Washed

human platelet suspensions (4Æ5· 108/ml) were

preincu-bated for 3 min in the presence or absence of magnesium sulphate (1Æ5 and 3Æ0 mmol/l) before the addition of collagen (1lg/ml). Six minutes after the addition of collagen, 2 mmol/l EDTA and 50lmol/l indomethacin were added to the reaction suspensions. The vials were then centrifuged for 3 min at 15 000 g. The thromboxane B2(TxB2) levels of

the supernatants were measured using an EIA kit according to the instructions of the manufacturer.

(3)

Determination of lactate dehydrogenase. Lactate dehydro-genase (LDH) was measured according to previously described methods (Wroblewski & Ladue, 1955). Platelets (4Æ5· 108

/ml) were preincubated with magnesium sul-phate (3Æ0 mmol/l) for either 10 or 30 min, followed by centrifugation 10 000 g for 5 min. An aliquot of superna-tant was incubated with phosphate buffer, containing 0Æ2 mg b-NADH, for 20 min at room temperature. There-after, 100ll of pyruvate solution was added and the absorbance wavelength was read at 334 nm using an ultra violet visible recording spectrophotometer (UV-160; Shimazu, Kyoto, Japan). A maximal value of LDH was constructed from sonicated platelets.

Measurement of protein kinase C activity. Washed human platelets (2· 109/ml) were incubated for 60 min at 37C

with phosphorus-32 (18Æ5 MBq/ml). Platelet suspensions were next washed twice with Tris-saline buffer. The [32

P]-labelled platelets were preincubated with magnesium sul-phate (1Æ5 mmol/l) in an aggregometer for 3 min; collagen (1lg/ml) was then added for 1 min to trigger protein kinase C activation. Activation was terminated by the addition of Laemmli sample buffer, and analysed by electrophoresis (12Æ5%; w/v) as described previously (Grabarek et al, 1992). The gels were dried, and the relative intensities of the radioactive bands were analysed using a Bio-imaging analyser system (FAL2000; Fuji, Tokyo, Japan) and were expressed as photostimulated luminescence/mm2 (PSL).

Estimation of platelet cyclic AMP and cyclic GMP formations. The method of Karniguian et al (1982) was followed. Briefly, platelet suspensions were warmed to 37C for 1 min, then PGE1(10lmol/l), nitroglycerin (10 lmol/l) or

magnesium sulphate (1Æ5 and 3Æ0 ml) was added and incubated for 6 min. The incubation was stopped and the solution was immediately boiled for 5 min. After cooling to 4C, the precipitated protein was collected as sediment after centrifugation. An aliquot (50ll) of supernatant was used to determine the cyclic AMP and cyclic GMP contents by EIA, following acetylation of the samples as described by the kit manufacturer.

Estimation of nitrate in human platelet suspensions. Platelet suspensions (1· 109/ml) were preincubated with

colla-gen (2lg/ml) and magnesium sulphate (1Æ5 and 3Æ0 mmol/l) for 6 min, followed by centrifugation (10 000 g) for 5 min. The supernatants were deprotei-nized by incubation with 95% ethanol at 4C for 30 min. The samples were then centrifuged for a further 7 min. It should be noted that the nitrate concentrations represent the total of both nitrite and nitrate concentrations in the platelet suspensions. This method reduced nitrate to NO via nitrite. The amount of nitrate in the platelet suspen-sions (10ll) was measured by adding a reducing agent (0Æ8% VCl3 in 1 mol/l HCl) to the purge vessel to convert

nitrate to NO which was stripped from the platelet suspensions by a helium purge gas. The NO was then drawn into a Sievers nitric oxide analyser (Sievers 280 NOA; Sievers, Boulder, CO, USA). Nitrate concentrations were calculated by comparison with standard solutions of sodium nitrate.

Statistical analysis. Experimental results are expressed as the means ± SEM and are accompanied by the number of observations. Data were assessed by analysis of variance (anova). If this analysis indicated significant differences among the group means, then each group was compared using the Newman–Keuls method. A P-value less than 0Æ05 was considered statistically significant.

RESULTS

Effect of magnesium sulphate on platelet aggregation in human platelet suspensions

Magnesium sulphate (0Æ6–3Æ0 mmol/l) dose-dependently inhibited platelet aggregation stimulated by collagen (1lg/ml), and thrombin (0Æ02 U/ml) in human platelets (Fig 1A and B). It also similarly inhibited ADP-induced (20lmol/l) platelet aggregation in the presence of fibrin-ogen (300lg/ml) (Fig 1B). Furthermore, magnesium

Fig 1. (A) Trace curves of magnesium sulphate on collagen (1lg/ml)-induced aggregation and (B) concentration–inhibition curve of magnesium sulphate on collagen- (1lg/ml, s), ADP-(20lmol/l, ,), thrombin- (0Æ02 U/ml, h) and arachidonic acid-(100lmol/l, e) induced platelet aggregation in washed human platelet suspensions. Platelets were preincubated with magnesium sulphate (0Æ6–3Æ0 mmol/l), agonists were then added to trigger aggregation (lower tracings) and ATP release (upper tracings) (A). Data are presented as a percentage of the control (means ± SEM, n¼ 6).

(4)

sulphate also inhibited the ATP release reaction when stimulated by agonists (i.e. collagen) (Fig 1A). The 50% inhibitory concentration (IC50) values of magnesium

sul-phate for platelet aggregation induced by ADP, collagen and thrombin were estimated to be approximately 1Æ7, 1Æ8 and 2Æ6 mmol/l respectively. On the other hand, magnesium sulphate (0Æ6–3Æ0 mmol/l) did not significantly inhibit arachidonic acid (100lmol/l) (Fig 1B) or the thromboxane A2analogue, U46619-induced (1lmol/l) platelet

aggrega-tion (data not shown). Furthermore, magnesium chloride (1Æ5 and 3Æ0 mmol/l) also showed a similar dose-dependent inhibitory effect on agonist-induced platelet aggregation (data not shown). In the following experiments, we used collagen as an agonist to explore the inhibitory mechanisms of magnesium sulphate in platelet aggregation.

Effect of magnesium sulphate on collagen-induced GPIIb/IIIa complex exposure in human platelets

Triflavin is an Arg-Gly-Asp-containing disintegrin from Trimeresurus flavoviridis snake venom (Sheu et al, 1996). Triflavin inhibits platelet aggregation through direct inter-ference with fibrinogen binding to the GPIIb/IIIa complex (Sheu et al, 1992). There is now a multitude of evidence suggesting that the binding of fibrinogen to the GPIIb/IIIa complex is the final common pathway for agonist-induced platelet aggregation. Therefore, we further evaluated whe-ther or not magnesium sulphate binds directly to the platelet GPIIb/IIIa complex, leading to inhibition of platelet aggregation induced by agonists.

In this study, the relative intensity of the fluorescence of FITC-triflavin (2lg/ml) bound directly to collagen-activated (1lg/ml) platelets was 88Æ5 ± 9Æ6 (n¼ 5) and was mark-edly reduced in the presence of 5 mmol/l EDTA (negative control, 17Æ5 ± 3Æ2, n¼ 5) (data not shown). At a concen-tration of 3Æ0 mmol/l, magnesium sulphate markedly inhibited FITC-triflavin binding to the GPIIb/IIIa complex in platelet suspensions (19Æ6 ± 3Æ8, n¼ 5) (data not shown), indicating that the mechanism of magnesium sulphate’s inhibitory effect on platelet aggregation may involve interference with fibrinogen binding to the GPIIb/ IIIa complex.

Effect of magnesium sulphate on phosphoinositide breakdown in human platelet suspensions

Phosphoinositide breakdown occurs in platelets activated by many different agonists (Broekman et al, 1980). We found that collagen (1lg/ml) induced the rapid formation of radioactive IP, IP2 and IP3 in human platelets loaded

with [3H]-inositol. We only measured [3H]-IP formation as an index of total inositol phosphate formation. As shown in Fig 2, the addition of collagen (1lg/ml) resulted in a rise of IP formation of about 2Æ3-fold compared with that in resting platelets [(5Æ8 ± 1Æ4 vs 13Æ5 ± 2Æ1)· 103

cpm]. In the presence of magnesium sulphate (1Æ5 and 3Æ0 mmol/l), the radioactivity of IP formation in colla-gen-stimulated human platelets markedly decreased. These results indicate that magnesium sulphate interferes with phosphoinositide breakdown in human platelets stimulated by collagen.

Effect of magnesium sulphate on [Ca2+]

imobilization

Free cytoplasmic Ca2+ concentrations in human platelets were measured by the Fura 2-AM loading method. In this study, collagen (1lg/ml) evoked an increase of [Ca2+]i from 32Æ8 ± 5Æ0 to 225Æ8 ± 39Æ2 nmol/l (data not shown), which was markedly inhibited in the presence of mag-nesium sulphate (1Æ5 mmol/l, 71Æ3 ± 2Æ4%; 3Æ0 mmol/l, 86Æ7 ± 10Æ1%; n¼ 4) (data not shown). This suggests that magnesium sulphate exerts an inhibitory effect on [Ca2+]i

mobilization in human platelets stimulated by collagen. Effect of magnesium sulphate on platelet membrane fluidity Platelet membrane fluidity was measured in DPH-labelled human platelets. Measurements using the fluorescent probe

Fig 2. Effect of magnesium sulphate on collagen-induced inositol monophosphate formation in human platelet suspensions. Platelets were labelled with [3H]-inositol and stimulated with or without

collagen (1lg/ml) in the presence of magnesium sulphate (1Æ5 and 3Æ0 mmol/l). Data are presented as the means ± SEM (n¼ 4). *P < 0Æ05 as compared with the resting group, #P < 0Æ05 as compared with the collagen group.

Fig 3. Fluorescence emission spectra of platelet membranes in the absence (A) or presence (B) of DPH (1lmol/l). Curves C and D are the emission spectra of membranes labelled with DPH in the pres-ence of magnesium sulphate (C) (1Æ5 mmol/l) and (D) (3Æ0 mmol/l) for 3 min. Profiles are representative examples of four similar experiments.

(5)

technique demonstrated that magnesium sulphate is capable of direct interaction with platelet membranes (Fig 3). Addition of magnesium sulphate (1Æ5 and 3Æ0 mmol/l) to platelet suspensions for 3 min resulted in a marked increase in DPH-related fluorescence intensity. This result implies that the inhibitory effect of magnesium sulphate on platelet aggregation may be due, at least in part, to the results of its effects on platelet membrane fluidity.

Effect of magnesium sulphate on TxB2formation

As shown in Table I, resting platelets produced relatively little TxB2compared with collagen-activated platelets. PGE1

(10lmol/l) inhibited TxB2formation in collagen-activated

platelets by 81% (data not shown). Furthermore, results obtained using various concentrations of magnesium sul-phate indicated that magnesium sulsul-phate (3Æ0 mmol/l) significantly inhibited TxB2formation in platelets stimulated

by collagen (1lg/ml) (Table I).

Effect of magnesium sulphate on LDH released from platelet cytosol

In this study, magnesium sulphate (3Æ0 mmol/l) treatment of human platelets for 10 min did not significantly increase LDH activity compared with resting platelets (resting platelets, 25Æ8 ± 3Æ4 units vs magnesium sulphate-treated platelets, 28Æ1 ± 3Æ9 units, n¼ 4), even when the incuba-tion time of magnesium sulphate with platelets was prolonged to 30 min (29Æ4 ± 3Æ3 units, n¼ 4) (data not shown). This indicates that although magnesium sulphate (1Æ5 and 3Æ0 mmol/l) significantly changes the fluidity of platelet membranes, by itself, it does not affect platelet permeabilization or induce platelet cytolysis under this range of concentrations.

Effect of magnesium sulphate on the formation of cyclic AMP, cyclic GMP and nitrate

The level of cyclic AMP in unstimulated platelets was low (31Æ5 ± 3Æ5 pmol/109platelets). Addition of PGE1

(10lmol/l) increased the cyclic AMP level to 236Æ7 ± 34Æ2 pmol/109platelets (Table II). When platelet suspensions were preincubated with magnesium sulphate (1Æ5 and 3Æ0 mmol/l), the cyclic AMP level increased to 77Æ1 ± 8Æ6 and 112Æ4 ± 12Æ8 pmol/109platelets

respect-ively (Table II). We also performed similar studies measur-ing the cyclic GMP response. The level of cyclic GMP in unstimulated platelets was very low, but when nitro-glycerin (10lmol/l) was added to the platelet suspensions, cyclic GMP levels increased from the resting level to 158Æ6 ± 13Æ7 pmol/109platelet (Table II). However, addi-tion of magnesium sulphate (1Æ5 and 3Æ0 lmol/l) resulted in no significant increase in platelet cyclic GMP levels (16Æ5 ± 1Æ5 and 16Æ0 ± 1Æ2 pmol/109platelets), even at a

higher concentration (5Æ0 lmol/l) (data not shown). On the other hand, NO was quantified using a sensitive and specific ozone redox-chemiluminescence detector. As shown in Table II, collagen (2Æ0 lg/ml) caused about a 2Æ9-fold rise in nitrate formation, compared with that in resting platelets. In the presence of magnesium sulphate (1Æ5 and 3Æ0 mmol/l), nitrate production did not significantly increase after incubation with platelets for 6 min (Table II).

Table I. Effect of magnesium sulphate on thromboxane B2

for-mation induced by collagen in washed human platelets.

Dose

Thromboxane B2(ng/ml)

Resting 45Æ8 ± 13Æ4 Collagen (lg/ml)

+Magnesium sulphate (mmol/l)

1 153Æ2 ± 19Æ4* 1Æ5 110Æ2 ± 12Æ7 3Æ0 85Æ5 ± 9Æ2 *P < 0Æ01 as compared with the resting group.

P < 0Æ05 as compared with the collagen group.

Platelet suspensions were preincubated with magnesium sul-phate (1Æ5 and 3Æ0 mmol/l) for 3 min at 37C and then collagen (1lg/ml) was added to trigger thromboxane B2formation. Data

are presented as the means ± SEM (n¼ 5).

Table II. Effect of magnesium sulphate on cyclic AMP, cyclic GMP and nitrate formation in washed human platelets.

Concentration Cyclic AMP (pmol/109platelets) Cyclic GMP (pmol/109platelets) Nitrate (lmol/l) Resting 31Æ5 ± 3Æ5 18Æ5 ± 2Æ1 4Æ8 ± 0Æ4 Prostaglandin E1(lmol/l) 10 236Æ7 ± 34Æ2* Nitroglycerin (lmol/l) 10 158Æ6 ± 13Æ7* Collagen (lg/ml) 2 13Æ8 ± 0Æ9*

Magnesium sulphate (mmol/l) 1Æ5 77Æ1 ± 8Æ6* 16Æ5 ± 1Æ5 5Æ9 ± 1Æ0 3Æ0 112Æ4 ± 12Æ8* 16Æ0 ± 1Æ2 5Æ6 ± 0Æ6 *P < 0Æ001 as compared with the resting groups.

Platelet suspensions were preincubated with magnesium sulphate (1Æ5 and 3Æ0 lmol/l) at 37C. Addition of prostaglandin E1, nitroglycerin and collagen in platelet suspensions served as a positive control of cyclic AMP, cyclic GMP and nitrate respectively. Data are presented as the means ± SEM (n¼ 5).

(6)

However, nitrate production did not increase even after prolongation of the incubation time to 30 min (data not shown). These results imply that the antiplatelet activity of magnesium sulphate may act at least partly through stimulation of the cyclic AMP, but not the NO/cyclic GMP pathway, in human platelets.

Effect of magnesium sulphate on collagen-stimulated phosphorylation of the 47 kDa protein

Stimulation of platelets with a number of different agonists induces activation of protein kinase C, which then phos-phorylates proteins of Mr 40 000–47 000 in addition to other proteins (Siess & Lapetina, 1989). In this study, phosphorylation was used to examine the role of magne-sium sulphate in activation of protein kinase C in human platelets. When collagen (1lg/ml) was added to human

platelets prelabelled with32PO

4for 2 min, a protein with an

apparent Mr of 47 000 (P47) was predominately phosphory-lated as compared with resting platelets (Fig 4A and B). On the other hand, magnesium sulphate (1Æ5 mmol/l) mark-edly inhibited the phosphorylation of P47 in human platelets stimulated by collagen (1lg/ml) (Fig 4). In this study, the extent of radioactivity in P47 was expressed as a relative detection density [photostimulated luminescence (PSL)/mm2] of the radioactive bands.

DISCUSSION

Magnesium sulphate is used widely to prevent seizures in pregnant women with hypertension (Lucas et al, 1995). The principal objective of this study was to describe the detailed mechanisms involved in the inhibition of agonist-induced human platelet aggregation by magnesium sul-phate. This inhibitory effect of magnesium sulphate was demonstrable with the use of various agonists: collagen, thrombin and ADP. The inhibition was directly propor-tional to the pharmacological concentrations of magnes-ium sulphate used. Lucas et al (1995) suggested that the magnesium sulphate regimen for pre-eclampsia consisted of an initial loading dose of 4 g of magnesium sulphate (i.v.) followed by a 10 g intramuscular dose and a maintenance dose of 5 g given intramuscularly every for 4 h. In general, the therapeutic concentration is considered to be between 2 and 4 mmol/l (Pritchard, 1979; Sibai et al, 1984). However, this is based on clinical experience and not directly related to the suppression of eclamptic convulsions. In this study, magnesium sulphate was employed at concentrations of about 0Æ6–3Æ0 mmol/l which inhibited platelet aggregation induced by agonists. This result indicates that the pharmacological concentra-tions of magnesium sulphate employed to inhibit platelet aggregation in vitro are reasonably close to those of blood concentrations obtained during a magnesium sulphate regimen in vivo. In this study, both platelet aggregation and the ATP release reaction induced by these agonists (i.e. collagen) appeared to be affected in the presence of magnesium sulphate. Therefore, this partly infers that magnesium sulphate may affect Ca2+ release from intra-cellular Ca2+ storage sites (i.e. dense tubular systems or dense bodies), and this is in accordance with the concept that intracellular Ca2+ release is responsible for the ATP release reaction (Charo et al, 1976). On the other hand, magnesium sulphate did not significantly inhibit U46619-induced (1lmol/l) or arachidonic acid-induced (100lmol/l) platelet aggregation, indicating that magnes-ium sulphate-mediated effects do not depend on the blockage of the arachidonic acid pathway.

Although the action mechanisms of various platelet aggregation agonists, such as collagen, ADP and thrombin, differ (Fig 5), magnesium sulphate significantly inhibited platelet aggregation that was stimulated by all of them. This implies that magnesium sulphate may block a common step shared by these inducers. These results also indicate that the site of action of magnesium sulphate is not at the receptor level of individual agonists. Triflavin acts by binding to the A

B

Fig 4. (A) Effect of magnesium sulphate on phosphorylation of a protein of Mr 47 000 (P47) in human platelets challenged with collagen. Platelets were preincubated with magnesium sulphate (1Æ5 mmol/l) before challenge with collagen (1 lg/ml). Lane 1, platelets with Tyrode’s solution; lane 2, platelets with collagen (1lg/ml); lane 3, platelets with magnesium sulphate (1Æ5 mmol/l) for 3 min followed by the addition of collagen (1lg/ml). The arrow indicates a protein of Mr 47 000 (P47). Data are representative examples of four similar experiments. (B) The relative detection densities of the radioactive bands are expressed as PSL/mm2. Data

(7)

GPIIb/IIIa complex on the platelet surface membrane, resulting in interference with the interaction of fibrinogen with its specific receptor (Sheu et al, 1992, 1996, 1999). In this study, we found that magnesium sulphate markedly inhibited FITC-triflavin binding to the GPIIb/IIIa complex (data not shown). Furthermore, Gawaz et al (1994) repor-ted that Mg+2 reduced fibrinogen binding to ADP- and collagen-stimulated platelets with an IC50 of about

3Æ0 mmol/l. It is well known that the fibrinogen–GPIIb/IIIa complex interaction is regulated by divalent cations, and that physiological concentrations of Ca+2and Mg+2support

fibrinogen binding (Gulino et al, 1992). At pharmacological levels, however, Mg+2may inhibit binding of fibrinogen to

the GPIIb/IIIa complex by altering the receptor conforma-tion. This might be caused by competition of Mg+2 with Ca+2ions for Ca+2binding sites in the GPIIb subunit (Gulino et al, 1992).

Conformational changes in the plasma membrane and/or changes in membrane fluidity represent a generally accep-ted mechanism for the antiplatelet effect of numerous drugs, including local anaesthetics, chlorpromazine and beta-blockers. Therefore, we wondered whether magnesium sulphate might also inhibit platelet aggregation by influen-cing membrane fluidity. To test this hypothesis, the fluor-escent probe, DPH, was used to label biological membranes. In this study, magnesium sulphate (1Æ5 and 3Æ0 mmol/l) markedly decreased the DPH-relative fluorescence intensity of platelet membranes. These findings suggest that changes

in platelet membrane fluidity may be the primary mechan-ism responsible for the antiplatelet effect of magnesium sulphate in vitro.

Furthermore, stimulation of platelets by agonists (i.e. collagen) results in phospholipase C-catalysed hydrolysis of the minor plasma membrane phospholipid, phosphatidyl-inositol 4,5-bisphosphate, with concomitant formation of inositol 1,4,5-trisphosphate and diacylglycerol (Fig 5). There is strong evidence that inositol 1,4,5-trisphosphate induces the release of Ca2+ from intracellular stores (Berridge, 1983). Diacylglycerol activates protein kinase C, inducing protein phosphorylation and a release reaction (Fig 5). In this study, phosphoinositide breakdown of collagen-activated platelets was inhibited by magnesium sulphate, suggesting that signal transductions of antiplatelet aggregation by magnesium sulphate is related to inhibition of phosphoinositide breakdown. TxA2 is an important

mediator of the release reaction and aggregation of platelets (Hornby, 1982). Collagen-induced formation of TxB2, a

stable metabolite of TxA2, was inhibited by magnesium

sulphate (3Æ0 mmol/l) (Table I). It has been demonstrated that phosphoinositide breakdown can induce TxA2

forma-tion via free arachidonic acid release by diglyceride lipase or by endogenous phospholipase A2 from membrane phos-pholipids (Fig 5) (McKean et al, 1981). Thus, it seems likely that phosphoinositide breakdown and TxB2formation play

a role in mediating the inhibitory effect of magnesium sulphate on human platelets.

Fig 5. Signal transductions of platelet aggregation. Agonists can activate several phospholipases, including phospholipase C (PLC) and phospholipase A2(PLA2). The products of the action of phospholipase Con phosphatidylinositol 4,5-bisphosphate (PIP2) include

1,2-diacyl-glycerol (DAG) and inositol 1,4,5-trisphosphate (IP3). DAG stimulates protein kinase C (PKC), followed by phosphorylation of a 47 kDa protein. IP3 induces the release of Ca+2from dense tubular systems (DTS). The major metabolite of arachidonic acid (AA) in platelets is thromboxane

A2(TxA2). Adenylate cyclase (AC) is activated by some prostaglandins (PGs) and adenosine, resulting in the hydrolysis of adenosine

tri-phosphate (ATP) to cyclic 3¢-5¢-adenosine monotri-phosphate (cyclic AMP). Cyclic 3¢-5¢-adenosine monotri-phosphate can activate cyclic AMP-dependent protein kinase (CAK), resulting in the inhibition of platelet activation.

(8)

The activation of human platelets is inhibited by two intracellular pathways, which are regulated by either cyclic AMP or cyclic GMP (Fig 5) (Walter et al, 1993). The importance of cyclic AMP in modulating platelet reactivity is well established (Karniguian et al, 1982). In addition to inhibiting most platelet responses, elevated levels of cyclic AMP decreased intracellular Ca2+ concentration by the uptake of Ca2+into the dense tubular system and negatively

affected the action of protein kinase C (Walter et al, 1993). In this study, we found that magnesium sulphate did not induce NO formation in human platelets. This result is in accordance with the cyclic GMP study, because the NO that is produced is biologically active, as most cellular actions of NO occur via stimulation of intracellular guanylate cyclase, leading to an increase in cyclic GMP (McDonald & Murad, 1996). Signalling by cyclic GMP somehow interferes with the agonist-stimulated phosphoinositide turnover that creates Ca2+-mobilizing second messengers and protein kinase C (McDonald & Murad, 1996). Therefore, the inhibitory effect of magnesium sulphate on collagen-induced phosphorylation of P47 and platelet aggregation seems to be mediated, at least partly, by an increase in cyclic AMP levels in human platelets.

In conclusion, the observations of this study suggest that magnesium sulphate inhibits agonist-induced human plate-let aggregation. This inhibitory effect may involve the following two mechanisms. (1) Magnesium sulphate may initially induce membrane fluidity changes on the platelet membrane, with a resulting interference of fibrinogen binding to the GPIIb/IIIa complex and activation of phospholipase C, followed by inhibition of phosphoinositide breakdown and thromboxane A2formation, thereby leading

to inhibition of both intracellular Ca2+ mobilization and

phosphorylation of P47. (2) Magnesium sulphate triggers the formation of cyclic AMP, which subsequently inhibits phosphoinositide breakdown and protein kinase C activity, finally resulting in inhibition of both the phosphorylation of P47 and intracellular Ca+2mobilization.

ACKNOWLEDGMENT

This work was supported by a grant from the National Science Council of Taiwan (NSC89-2320-B-038–002-M53). REFERENCES

Berridge, M.J. (1983) Rapid accumulation of inositol trisphosphate reveals that agonists hydrolyse polyphosphoinositides instead of phosphatidylinositol. Biochemical Journal, 212, 249–258. Born, G.V.R. & Cross, M.J. (1963) The aggregation of blood

plate-lets. Journal of Physiology, 168, 178–195.

Broekman, M.J., Ward, J.W. & Marcus, A.J. (1980) Phospholipid metabolism in phosphatidyl inositol, phosphatic acid and lyso-phospholipids. Journal of Clinical Investigation, 66, 275–283. Charo, I.F., Feinman, R.D. & Detwiler, T.C. (1976) Inhibition of

platelet secretion by an antagonist of intracellular calcium. Bio-chemical and Biophysical Research Communication, 72, 1462–1467. Gawaz, M., Ott, I., Reininger, A.J. & Neumann, F.J. (1994) Effect of magnesium on platelet aggregation and adhesion. Thrombosis and Haemostasis, 72, 912–918.

Grabarek, J., Raychowdhury, M., Ravid, K., Kent, K.C., Newman, P.J. & Ware, J.A. (1992) Identification and functional char-acterization of protein kinase C isozymes in platelets and HEL cells. Journal of Biological Chemistry, 267, 10011–10017. Grynkiewicz, G., Poenie, M. & Tsien, R.Y. (1985) A new generation

of Ca2+induces with greatly improved fluorescence properties.

Journal of Biological Chemistry, 260, 3440–3450.

Gulino, D., Boudignon, C., Zhang, L., Concord, E., Rabiet, M.J. & Marguerie, G. (1992) Ca+2-binding properties of the platelet

glycoprotein IIb ligand-interacting domain. Journal of Biological Chemistry, 267, 11001–11007.

Hass, W.K., Easton, J.D., Adams, H.P., Pryse-Philips, W., Molony, B.A., Anderson, S. & Kamm, B. (1989) A randomized trial comparing ticlopidine hydrochloride with aspirin for the pre-vention of stroke in high-risk patients. The New England Journal of Medicine, 321, 501–507.

Hornby, E.J. (1982) Evidence that prostaglandin endoperoxides can induce platelet aggregation in the absence of thromboxane A2

production. Biochemical Pharmacology, 31, 1158–1160. Huang, T.F., Sheu, J.R. & Teng, C.M. (1991a) A potent antiplatelet

peptide, triflavin, from Trimeresurus flavoviridis snake venom. Biochemical Journal, 277, 351–357.

Huang, T.F., Sheu, J.R. & Teng, C.M. (1991b) Mechanism of action of a potent antiplatelet peptide, triflavin, from Trimeresurus flavo-viridis snake venom. Thrombosis and Haemostasis, 66, 489–493. Hwang, D.L., Yen, C.F. & Nadler, J.L. (1992) Effect of intracellular

magnesium on platelet activation and intracellular calcium mobilization. American Journal of Hypertension, 5, 700–706. Karniguian, A., Legrand, Y.J. & Caen, J.P. (1982) Prostaglandins:

specific inhibition of platelet adhesion to collagen and relation-ship with c AMP level. Prostaglandin, 23, 437–457.

Kitagawa, S., Shinohara, T. & Kametani, F. (1984) Effects of alco-hols on ADP-induced aggregation and membrane fluidity of gel-filtered bovine blood platelets. Journal of Membrane Biology, 79, 97–102.

Lucas, M.J., Leveno, K.J. & Cunningham, F.G. (1995) A comparison of magnesium sulfate with phenytoin for the prevention of eclampsia. The New England Journal of Medicine, 333, 201–205. McDonald, L.J. & Murad, F. (1996) Nitric oxide and cyclic GMP

signaling. Proceedings of the Society for Experimental Biology and Medicine, 211, 1–6.

McKean, M.L., Smith, J.B. & Silver, W.J. (1981) Formation of lysophosphatidylcholine in human platelets in response to thrombin. Journal of Biological Chemistry, 256, 1522–1524. Nadler, J.L., Goodson, S. & Rude, R.K. (1987) Evidence that

pros-tacyclin mediates the vascular action of magnesium in humans. Hypertension, 9, 379–383.

Nadler, J., Malayan, S. & Luong, H. (1992) Evidence that intracellular free magnesium deficiency plays a key role in increased platelet reactivity in type II diabetes mellitus. Diabetes Care, 15, 835–841.

Pritchard, J.A. (1979) The use of magnesium sulfate in pre-eclampsia and pre-eclampsia. Journal of Reproductive Medicine, 23, 107–114.

Rasmussen, H.S., McNair, P., Goransson, L., Balslov, S., Larsen, O.G. & Aurup, P. (1988) Magnesium deficiency in patients with and without acute myocardial infarction uncovered by an intravenous loading test. Archives of Internal Medicine, 148, 329– 332.

Ravn, H.B., Vissinger, H., Kristensen, S.D. & Husted, S.E. (1996a) Magnesium inhibits platelet activity: an in vitro study. Thrombosis and Haemostasis, 76, 88–93.

Ravn, H.B., Vissinger, H., Kristensen, S.D., Wennmalm, A., Thygesen, K. & Husted, S.E. (1996b) Magnesium inhibits platelet

(9)

activity: an infusion study in healthy volunteers. Thrombosis and Haemostasis, 75, 939–944.

Shechter, M., Sharir, M., Paul Labrador, M.J., Forrester, J., Silver, B. & Merz, N.B. (2000) Oral magnesium therapy improves endothelial function in patients with coronary artery disease. Circulation, 102, 2353–2358.

Sheu, J.R., Teng, C.M. & Huang, T.F. (1992) Triflavin, an RGD-containing antiplatelet peptide, binds to GP IIIa of ADP-stimulated platelets. Biochemical and Biophysical Research Communication, 189, 1236–1242.

Sheu, J.R., Chao, S.H., Yen, M.H. & Huang, T.F. (1994) In vivo antithrombotic effect of triflavin, an Arg-Gly-Asp containing peptide on platelet plug formation in mesenteric microvessels of mice. Thrombosis and Haemostasis, 72, 617–621.

Sheu, J.R., Lin, C.H., Peng, C.H. & Huang, T.F. (1996) Triflavin, an Arg-Gly-Asp-containing peptide, inhibits the adhesion of tumor cells to matrix protein via binding to multiple integrin receptors expressed on human hepatoma cells. Proceedings of the Society for Experimental Biology and Medicine, 213, 71–79.

Sheu, J.R., Hung, W.C., Wu, C.H., Ma, M.C., Kan, Y.C., Lin, C.H., Lin, M.S., Luk, H.N. & Yen, M.H. (1999) Reduction in

lipopoly-saccharide-induced thrombocytopenia by triflavin in a rat model of septicemia. Circulation, 99, 3056–3062.

Sibai, B.M., Graham, J.M. & McCubbin, J.H. (1984) A comparison of intravenous and intramuscular magnesium sulfate regimens in pre-eclampsia. American Journal of Obstetrics and Gynecology, 150, 728–733.

Siess, W. & Lapetina, E.G. (1989) Platelet aggregation induced by alpha 2-adrenoceptor and protein kinase C activation. A novel synergism. Biochemical Journal, 263, 377–385.

Walter, U., Eigenthaler, M., Geiger, J. & Reinhard, M. (1993) Role of cyclic nucleotide-dependent protein kinases and their common substrate VASP in the regulation of human platelets. Advances in Experimental Medicine and Biology, 344, 237–249.

Watson, K.L., Moldow, L.F., Ogburn, P.L. & Jacob, H.S. (1986) Magnesium sulfate: rationale for its use in preeclampsia. Pro-ceedings of the National Academy of Sciences of the United States of America, 83, 1075–1078.

Wroblewski, F. & Ladue, J.S. (1955) Lactic dehydrogenase activity in blood. Proceedings of the Society for Experimental Biology and Medicine, 90, 210–215.

數據

Fig 3. Fluorescence emission spectra of platelet membranes in the absence (A) or presence (B) of DPH (1 lmol/l)
Table II. Effect of magnesium sulphate on cyclic AMP, cyclic GMP and nitrate formation in washed human platelets.
Fig 4. (A) Effect of magnesium sulphate on phosphorylation of a protein of Mr 47 000 (P47) in human platelets challenged with collagen
Fig 5. Signal transductions of platelet aggregation. Agonists can activate several phospholipases, including phospholipase C (PLC) and phospholipase A 2 (PLA2)

參考文獻

相關文件

The first row shows the eyespot with white inner ring, black middle ring, and yellow outer ring in Bicyclus anynana.. The second row provides the eyespot with black inner ring

The underlying idea was to use the power of sampling, in a fashion similar to the way it is used in empirical samples from large universes of data, in order to approximate the

• helps teachers collect learning evidence to provide timely feedback &amp; refine teaching strategies.. AaL • engages students in reflecting on &amp; monitoring their progress

Robinson Crusoe is an Englishman from the 1) t_______ of York in the seventeenth century, the youngest son of a merchant of German origin. This trip is financially successful,

fostering independent application of reading strategies Strategy 7: Provide opportunities for students to track, reflect on, and share their learning progress (destination). •

Strategy 3: Offer descriptive feedback during the learning process (enabling strategy). Where the

How does drama help to develop English language skills.. In Forms 2-6, students develop their self-expression by participating in a wide range of activities

Now, nearly all of the current flows through wire S since it has a much lower resistance than the light bulb. The light bulb does not glow because the current flowing through it