You are on page 1of 4

Clinical and Experimental Pharmacology and Physiology (2007) 34, 1272–1275 doi: 10.1111/j.1440-1681.2007.04720.

Blackwell
Original Publishing
Articles al. of Asia

EFFECT OF THE HEME OXYGENASE INDUCER HEMIN ON BLOOD


GY
Anticoagulant
Rochefort et
effect hemin assessed by US

HAEMOSTASIS MEASURED BY HIGH-FREQUENCY ULTRASOUND

Gaël Y Rochefort,* Rachel Libgot,† Nicolas Desbuards,* Deborah Schlecht,* Jean-Michel Halimi,*
Frederic Ossant,† Veronique Eder* and Daniel Antier*
*LAB.P.ART. EA3852 IFR135 and †LUSSI FRE CNRS 2448, François Rabelais University, Faculty of Medicine, Tours, France

SUMMARY effect.4,7,10 Heme arginate does not affect coagulation, whereas hae-
matin has often been associated with coagulopathy and occasional
1. Heme compounds, like hemin, a heme oxygenase-1 inducer, bleeding.11,12 A degradation product or derivative of haematin, namely
are used in the treatment of acute porphyria treatment. The haematin-derived anticoagulant, was found to induce prolongation
side-effects of hemin on haemostasis have been reported. To of clotting times in vitro.13 Indeed, hemin has been associated with
address those effects, in the present study we used a sensitive, defects in haemostasis characterized by elevation of prothrombin
high-frequency ultrasound technique to record acoustic velocity time, partial thromboplastin time and thrombin time,5 and it has been
and to investigate whole blood clotting in Wistar rats treated proven to suppress haemorrhagic shock.14 Therefore, hemin was
chronically with hemin (50 mg/kg per day). hypothesized to exert its anticoagulant effects by inactivating
2. The hemin-induced disturbances in haemostasis measured thrombin.5 Conversely, hemin has been associated with no adverse
were comparable to the heparin reference treatment, with a sig- effects on haemostasis in healthy volunteers.3
nificant decrease in clotting velocity in both groups compared Hemin can also cause liver injury, leading to coagulation disorder.
with controls (e.g. the time to clot was 40 ± 5, 53 ± 13 and However, You et al. performed a toxicity study in rats treated with high
10 ± 2 min, respectively; P < 0.05). Precautions must be taken doses of hemin (93, 168 and 300 mg/kg per day) and reported no
when using high doses of hemin or in the treatment of bleeding liver or renal dysfunction.15 Odaka et al. reported that 30 mmol/kg,
diseases. s.c., (i.e. 20 mg/kg) hemin treatment induced heme oxygenase-1
3. Further investigations are required to explore the effects (HO-1) and prevented halothane-induced hepatotoxicity.16 Similarly,
of hemin in thrombosis models, because it could be a promising Chiu et al. showed that 30 mmol/kg, i.p., hemin may be hepato-
‘old drug’ for the treatment of venous thrombosis in patients. protective in rats because HO-1 is suggested to be an important com-
Key words: haemostasis, hemin, high-frequency ultrasound, ponent of the anti-oxidant defence during hepatotoxicity.17 Finally,
rat. Morio et al. reported that hemin treatment with a dose of 50 mmol/kg
(i.e. 35 mg/kg) had minimal effects on the liver, suggesting that HO-1
may play a limited role in hepatotoxicity.18
INTRODUCTION Clinical assessment of the blood clotting mechanism is usually
made by measuring the time necessary for a plasma sample to clot.
Heme compounds are the most efficient in the treatment of acute
Coagulation, clot formation and thrombosis are already assessed
porphyria,1,2 a well-defined autonomic dominant disorder of heme
using non-invasive processes, such as ultrasound techniques.19–21
biosynthesis with a genetic frequency of 1 : 10 000–1 : 50 000 and
During the clotting process, specific changes in several ultrasound
characterized by acute life-threatening attacks of non-specific
parameters, such as velocity, attenuation coefficient and backscatter
neurological symptoms.3–6 Given intravenously at moderate doses
coefficient, can be recorded quantitatively.22–24 Thus, high-frequency
(3– 4 mg/kg per day), heme compounds usually result in a rapid
ultrasound techniques have been evaluated and compared with
clinical improvement, often within 1–2 days, especially when therapy
reference methods (i.e. the manual tilt tube and the fibrometer) and
is started early in an attack.4,7,8 Such compounds are now available as
are validated for clinical use, allowing a highly sensitive description
heme arginate (Normosang®; Orphan Europe, Puteaux, France) and
of the clotting process.20,25
as haematin (Panhematin; Ovation Pharmaceuticals, Deerfield, IL,
The aims of the present study were to determine the anticoagulant
USA), which is a lyophilized powder of hemin. Haematin infusions
effect of hemin using in vitro high-frequency ultrasound analysis of
have also been reported to be a safe and useful therapeutic modality
whole blood clotting and to evaluate the haemostatic effect of a high
to maintain remission of allograft erythropoietic protoporphyria and
dose of hemin (50 mg/kg per day).
to prevent further recurrence of protoporphyric graft disease.9
In addition to therapeutic properties, side-effects of hemin have
been identified, including haemolysis and a transient anticoagulant METHODS
Experiments were performed in accordance with the Guidelines for the Care
and Use of Laboratory Animals published by the US National Institutes of
Correspondence: Daniel Antier, LABPART–EA 3852 10 Bd Tonnellé BP Health (NIH Publication no. 85–23, revised 1996; http://grants1.nih.gov/
3223, 37032 Tours Cedex 1, France. Email: antier@med.univ-tours.fr grants/olaw/GuideBook.pdf ), with European Directives (86/609/CEE; http://
Received 30 December 2006; revision 16 April 2007; accepted 13 May 2007. eur-lex.europa.eu/LexUriServ/site/en/com/2001/com2001_0703en01.pdf ),
© 2007 The Authors in accordance with the regulations of the official edict of the French Ministry
Journal compilation © 2007 Blackwell Publishing Asia Pty Ltd of Agriculture and approved by the local ethics committee.
Anticoagulant effect of hemin assessed by US 1273

Animals, treatment and sampling Statistical analysis


Male Wistar rats (approximately 350 g; Janvier, Le Genest-saint-Isles, France) All data are expressed as the mean±SEM. Differences between control and
were chronically injected with 50 mg/kg per day, i.p., hemin in 0.2 mL hemin-treated groups were analysed with the Mann–Whitney test. Overall
dimethylsulphoxide (DMSO; both from Sigma, Saint-Quentin Fallavier, statistical significance was set at P < 0.05.
France; n = 5) for 7 days. Untreated (control; n = 6) and heparin-treated
(500 IU/kg per day; n = 5) rats were used as a reference. After the 7 day treat-
ment and 2 h after the last injection, whole blood samples were collected RESULTS
into 3.5 mL vacuum citrated tubes (3.2%) and heated at 37.0 ± 0.1∞C. The
coagulation process was initiated by the addition of calcium chloride (100 mL Serum heme levels were either undetectable or < 1 mmol/L in all rats
of a 0.5 mol/L solution) and acoustic measurements were recorded in a blood before treatment, but serum heme levels increased significantly
chamber within 30 min after sampling, in an incubator adjusted at 37.0 ± (n = 5; P < 0.005) to 59.6 ± 9.9 mmol/L after 1 week of daily treat-
0.1∞C, as described previously.20,21 ment with 50 mg/kg per day hemin.
Mean acoustic velocities as a function of time measured for
High-frequency ultrasound analysis untreated controls, heparin-treated (reference) and 50 mg/kg per day
hemin-treated rats were recorded and the results are shown in Fig. 1.
A 45 MHz focused transducer, with a broadband (200 MHz) pulser/receiver Indeed, the clotting process for 50 mg/kg per day hemin-treated rats
(both designed by Panametrics, La Garenne Colombes, France), was set on is comparable to that seen in heparin-treated animals.
a measurement cell in order to adjust the focal depth of the transducer to
Mean values for time t0, slope S and the final value I for the three
the middle of the blood chamber propagation path. The technique used to
measure acoustic velocity (m/s), and described elsewhere,20,21 was a classical groups are shown in Fig. 2. Whole blood from control rats began to
differential pulse echo method. Briefly, the time interval between ultrasound
pulses from the top and bottom of the blood chamber was recorded. Vari-
ations in the interval time were due to changes in the velocity in the blood
sample and allowed estimation of the blood acoustic velocity of the sample.

Recorded parameters
Acoustic velocities were digitized at 500 MHz with 8-bit resolution, recorded
with a 15 s time resolution and expressed as a function of the experiment
time.
For each blood samples, three characteristics were extracted: (i) the time,
t0, from which the acoustic velocity increased strongly, which corresponds
to serum expulsion and the beginning of blood clotting;20,21 (ii) the slope, S,
of the increase in acoustic velocity, which is related to the transition from
liquid to gel; and (iii) the final value, I, of the velocity at t = 100 min, which
is an index of clot density.

Measurement of serum hemin concentrations


To obtain an indirect measure of hemin metabolism after daily treatment, Fig. 1 Whole blood acoustic velocity during the clotting process. Mean
serum concentrations of heme, which is one of the metabolic products of (±SEM) acoustic velocities (in m/s) as a function of time (in min) measured
hemin, were determined, as described previously,26 prior to drug injection for untreated controls (), 500 IU/kg per day heparin ()- and 50 mg/kg
and at the end of the study. per day hemin ()-treated rats.

Fig. 2 Whole blood ultrasound parameters during the clotting process. Mean values for (a) time t0 (in min), (b) slope S (in m/s2) and (c) final value I (in
m/s) for untreated controls (), 500 IU/kg per day heparin ()- and 50 mg/kg per day hemin ( )-treated rats. *P < 0.05 versus untreated controls.

© 2007 The Authors


Journal compilation © 2007 Blackwell Publishing Asia Pty Ltd
1274 GY Rochefort et al.

clot early (low t0), rapidly (high S ) and completely (high I), whereas 2. Pierach CA. Haem and porphyria attacks. Lancet 1989; ii: 213 –14.
whole blood from heparin- and 50 mg/kg per day hemin-treated rats 3. Volin L, Rasi V, Vahtera E, Tenhunen R. Heme arginate: Effects on
showed significant (P < 0.05) increases in t0 and significant hemostasis. Blood 1988; 71: 625–8.
4. Anderson KE, Bloomer JR, Bonkovsky HL et al. Recommendations
decreases in S and I.
for the diagnosis and treatment of the acute porphyrias. Ann. Intern.
Med. 2005; 142: 439–50.
5. Glueck R, Green D, Cohen I, Ts’ao CH. Hematin: Unique effects of
DISCUSSION
hemostasis. Blood 1983; 61: 243–9.
After 1 week of daily treatment with 50 mg/kg per day hemin, serum 6. Hift RJ, Meissner PN. An analysis of 112 acute porphyric attacks in
heme levels were significantly increased compared with concentra- Cape Town, South Africa: Evidence that acute intermittent porphyria
and variegate porphyria differ in susceptibility and severity. Medicine
tions before treatment, confirming that the i.p. hemin treatment was
2005; 84: 48–60.
efficient and reproducible. 7. Anderson KE, Bonkovsky HL, Bloomer JR, Shedlofsky SI. Reconsti-
We used a highly sensitive ultrasound technique to record the tution of hematin for intravenous infusion. Ann. Intern. Med. 2006; 144:
blood clotting process of animals treated or not with 50 mg/kg per 537–8.
day, i.p. hemin. Intraperitoneal injections were used to avoid general 8. Suarez JI, Cohen ML, Larkin J, Kernich CA, Hricik DE, Daroff RB.
anaesthesia and bolus effects, which are often observed after i.v. Acute intermittent porphyria: Clinicopathologic correlation. Report of
injections. All parameters recorded indicated that hemin, adminis- a case and review of the literature. Neurology 1997; 48: 1678 – 83.
9. Dellon ES, Szczepiorkowski ZM, Dzik WH et al. Treatment of recur-
tered at a high dose, causes disturbances in haemostasis, as does
rent allograft dysfunction with intravenous hematin after liver trans-
heparin. Thus, the discrepancies reported in the literature3,5,11 could plantation for erythropoietic protoporphyria. Transplantation 2002; 73:
be explained by individual fluctuations or clinical history. Further- 911–15.
more, lower doses of hemin could induce limited haemostatic effects, 10. Daimon M, Susa S, Igarashi M, Kato T, Kameda W. Administration
as suggest previously by Maeshima et al.14 regarding the ability of heme of heme arginate, but not hematin, caused anaphylactic shock. Am. J.
arginate to induce HO-1, the rate-limiting enzyme in heme catabo- Med. 2001; 110: 240.
lism involved in the inhibition of platelet-dependent thrombosis.27 11. Green D, Ts’ao CH. Hematin: Effects on hemostasis. J. Lab. Clin. Med.
1990; 115: 144–7.
Heme oxygenase-1 is ubiquitously distributed and strongly induced
12. Mustajoki P, Nordmann Y. Early administration of heme arginate for
by oxidative, nitrosative, osmotic and haemodynamic stresses.28 acute porphyric attacks. Arch. Intern. Med. 1993; 153: 2004 –8.
Induction of HO-1 results in the catabolism of the pro-oxidant heme 13. Jones RL. Hematin-derived anticoagulant. Generation in vitro and in
to bile pigments, biliverdin and bilirubin, which are potent anti- vivo. J. Exp. Med. 1986; 163: 724–39.
oxidants capable of scavenging peroxy radicals and inhibiting lipid 14. Maeshima K, Takahashi T, Uehara K et al. Prevention of hemorrhagic
peroxidation, providing an important cellular defence mechanism shock-induced lung injury by heme arginate treatment in rats. Biochem.
Pharmacol. 2005; 69: 1667–80.
against tissue injury.29 Moreover, HO-1 is responsible for the liber-
15. You KS, Wu B, Huang ZQ et al. Effect of hemin in treating hemorrhagic
ation of the diatomic gas CO, which has significant cytoprotective anemia and toxicity. Zhongguo Yao Li Xue Bao 1996; 17: 284 – 6 (in
effects, including anti-inflammatory,29–32 antiproliferative,33,34 anti- Chinese).
apoptotic35,36 and anticoagulating activity.37 16. Odaka Y, Takahashi T, Yamasaki A et al. Prevention of halothane-
Dimethylsulphoxide, the vehicle used in the present study for induced hepatotoxicity by hemin pretreatment: Protective role of heme
hemin, has been reported to be a modulator of procoagulant and oxygenase-1 induction. Biochem. Pharmacol. 2000; 59: 871– 80.
fibrinolytic activities, as well as metastatic ability of B16 melanoma 17. Chiu H, Brittingham JA, Laskin DL. Differential induction of heme
oxygenase-1 in macrophages and hepatocytes during acetaminophen-
cells when used at a concentration of 0.2% in vitro.38 Moreover,
induced hepatotoxicity in the rat: Effects of hemin and biliverdin.
experimental data have shown that 5% DMSO decreases the Toxicol. Appl. Pharmacol. 2002; 181: 106–15.
adhesiveness of blood platelets in vitro and that DMSO may be a 18. Morio LA, Leone A, Sawant SP et al. Hepatic expression of heme
good antagonist against platelet aggregation, as well as thrombus oxygenase-1 and antioxidant response element-mediated genes follow-
formation, in vivo.39 These effects of DMSO were likely to be ing administration of ethinyl estradiol to rats. Toxicol. Appl. Pharmacol.
involved in its protective effect against ischaemic injury40,41 when 2006; 216: 416–25.
used at high doses (1–30%). In the present study, the final concen- 19. Huang CC, Wang SH, Tsui PH. Detection of blood coagulation and
clot formation using quantitative ultrasonic parameters. Ultrasound
tration of DMSO injected into rats was low (approximately 0.05%,
Med. Biol. 2005; 31: 1567–73.
v/v). Moreover, DMSO has no effect on the inhibition of thrombus 20. Libgot R, Ossant F, Gruel Y, Lermusiaux P, Patat F. High frequency
formation in vivo in mice.42 ultrasound characterization of the blood clotting process. Intra- and
In conclusion, hemin is known to be effective and well tolerated inter-individual variations. IEEE Ultrasonics Symp. 2005; 4: 2259 –
in the treatment of acute porphyria. High doses of hemin could cause 62.
disturbances in haemostasis, thus bleeding diseases and surgery 21. Ossant F, Libgot R, Coupé P, Lermusiaux P, Patat F. High frequency
ultrasound characterization of the coagulation process of whole blood.
should be relative contraindications to the use of hemin, especially
IEEE Ultrasonics Symp. 2004; 2: 846–9.
in the case of concomitant heparin, and thus precautions must be 22. Shung KK, Fei DY, Ballard 3rd JO. Further studies on ultrasonic pro-
taken when using high doses of hemin. Finally, our results focus on perties of blood clots. J. Clin. Ultrasound 1986; 14: 269–75.
major potential perspectives for hemin in the treatment of thrombo- 23. Shung KK, Fei DY, Yuan YW, Reeves WC. Ultrasonic characterization
embolic diseases. of blood during coagulation. J. Clin. Ultrasound 1984; 12: 147–53.
24. Voleisis A, Kazys R, Mazeika L, Sliteris R, Voleisiene B, Grybauskas P.
Ultrasonic method for the whole blood coagulation analysis. Ultrasonics
REFERENCES 2002; 40: 101–7.
25. Machado JC, von Kruger MA, Fontes EM, de Almeida MM. Evaluation
1. Peters HA, Levine R, Cripps D. Acute intermittent porphyria. Neuro- of an ultrasonic method applied to the measurement of blood coagu-
logy 1998; 50: 1932 – 3. lation time. Physiol. Meas. 1997; 18: 129–43.

© 2007 The Authors


Journal compilation © 2007 Blackwell Publishing Asia Pty Ltd
Anticoagulant effect of hemin assessed by US 1275

26. Shamloul R, Wang R. Monitoring circulatory heme level in hemin 35. Brouard S, Otterbein LE, Anrather J et al. Carbon monoxide generated
therapy for lowering blood pressure in rats. Cell Mol. Biol. 2005; 51: by heme oxygenase 1 suppresses endothelial cell apoptosis. J. Exp.
507–12. Med. 2000; 192: 1015–26.
27. Peng L, Mundada L, Stomel JM et al. Induction of heme oxygenase- 36. Silva G, Cunha A, Gregoire IP, Seldon MP, Soares MP. The anti-
1 expression inhibits platelet-dependent thrombosis. Antioxid. Redox apoptotic effect of heme oxygenase-1 in endothelial cells involves the
Signal. 2004; 6: 729 – 35. degradation of p38 alpha MAPK isoform. J. Immunol. 2006; 177:
28. Maines MD. The heme oxygenase system: A regulator of second mes- 1894–903.
senger gases. Annu. Rev. Pharmacol. Toxicol. 1997; 37: 517–54. 37. Brune B, Ullrich V. Inhibition of platelet aggregation by carbon mon-
29. Morita T. Heme oxygenase and atherosclerosis. Arterioscler. Thromb. oxide is mediated by activation of guanylate cyclase. Mol. Pharmacol.
Vasc. Biol. 2005; 25: 1786 –95. 1987; 32: 497–504.
30. Otterbein LE, Bach FH, Alam J et al. Carbon monoxide has anti- 38. Casali B, Lampugnani MG, Riganti M et al. DMSO-induced changes
inflammatory effects involving the mitogen-activated protein kinase in the procoagulant and fibrinolytic activity of B16 melanoma cells:
pathway. Nat. Med. 2000; 6: 422 – 8. Influence on lung colony formation. Clin. Exp. Metastasis 1988; 6:
31. Wagener FA, Volk HD, Willis D et al. Different faces of the heme- 377–85.
heme oxygenase system in inflammation. Pharmacol. Rev. 2003; 55: 39. Gorog P, Kovacs IB. Antiarthritic and antithrombotic effects of topi-
551–71. cally applied dimethyl sulfoxide. Ann. N.Y. Acad. Sci. 1975; 243: 91– 7 .
32. Wunder C, Potter RF. The heme oxygenase system: Its role in liver 40. Jacob SW, Herschler R. Pharmacology of DMSO. Cryobiology 1986;
inflammation. Curr. Drug Targets Cardiovasc. Haematol. Disord. 23: 14–27.
2003; 3: 199–208. 41. Saeed SA, Karimi SJ, Suria A. Differential effects of dimethyl sulfoxide
33. Morita T, Mitsialis SA, Koike H, Liu Y, Kourembanas S. Carbon mon- on human platelet aggregation and arachidonic acid metabolism.
oxide controls the proliferation of hypoxic vascular smooth muscle Biochem. Med. Metab. Biol. 1988; 40: 143–50.
cells. J. Biol. Chem. 1997 ; 272: 32 804 – 9. 42. Lindenblatt N, Bordel R, Schareck W, Menger MD, Vollmar B. Vas-
34. Duckers HJ, Boehm M, True AL et al. Heme oxygenase-1 protects against cular heme oxygenase-1 induction suppresses microvascular thrombus
vascular constriction and proliferation. Nat. Med. 2001; 7: 693–8. formation in vivo. Arterioscler. Thromb. Vasc. Biol. 2004; 24: 601– 6.

© 2007 The Authors


Journal compilation © 2007 Blackwell Publishing Asia Pty Ltd

You might also like