You are on page 1of 16

Basic & Clinical Pharmacology & Toxicology, 2015, 116, 398–413 Doi: 10.1111/bcpt.

12335

Anti-inflammatory Effect of Rosmarinic Acid and an Extract of


Rosmarinus officinalis in Rat Models of Local and Systemic
Inflammation
Joao Rocha1, Maria Eduardo-Figueira1, Andreia Barateiro1, Adelaide Fernandes1, Dora Brites1, Rosario Bronze2, Catarina MM Duarte2,
Ana Teresa Serra2, Rui Pinto1, Marisa Freitas3, Eduarda Fernandes3, Beatriz Silva-Lima1, Helder Mota-Filipe1 and Bruno Sepodes1
1
iMed.ULisboa, Faculty of Pharmacy - University of Lisbon, Lisboa, Portugal, 2ITQB/IBET, Quinta do Marqu^es Estacß~ao Agronomica Nacional,
Oeiras, Portugal and 3REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto,
Porto, Portugal
(Received 14 July 2014; Accepted 25 September 2014)

Abstract: Rosmarinic acid is a polyphenolic compound and main constituent of Rosmarinus officinalis and has been shown to
possess antioxidant and anti-inflammatory properties. We aimed to evaluate the anti-inflammatory properties of rosmarinic acid
and of an extract of R. officinalis in local inflammation (carrageenin-induced paw oedema model in the rat), and further evaluate
the protective effect of rosmarinic acid in rat models of systemic inflammation: liver ischaemia–reperfusion (I/R) and thermal
injury models. In the local inflammation model, rosmarinic acid was administered at 10, 25 and 50 mg/kg (p.o.), and the extract
was administered at 10 and 25 mg/kg (equivalent doses to rosmarinic acid groups) to male Wistar rats. Administration of ros-
marinic acid and extract at the dose of 25 mg/kg reduced paw oedema at 6 hr by over 60%, exhibiting a dose–response effect,
suggesting that rosmarinic was the main contributor to the anti-inflammatory effect. In the liver I/R model, rosmarinic acid was
administered at 25 mg/kg (i.v.) 30 min. prior to the induction of ischaemia and led to the significant reduction in the serum con-
centration of transaminases (AST and ALT) and LDH. In the thermal injury model, rosmarinic acid was administered at 25 mg/
kg (i.v.) 5 min. prior to the induction of injury and significantly reduced multi-organ dysfunction markers (liver, kidney, lung)
by modulating NF-jB and metalloproteinase-9. For the first time, the anti-inflammatory potential of rosmarinic acid has been
identified, as it causes a substantial reduction in inflammation, and we speculate that it might be useful in the pharmacological
modulation of injuries associated to inflammation.

Rosmarinus officinalis L., popularly named rosemary, has It has been widely recognized for many years that certain
been used in folk medicine with several pharmacological types of inflammatory tissue injury are mediated by reactive
effects being associated to its consumption, including its anti- oxygen metabolites and that in addition to promoting direct
inflammatory effects [1,2], and rosmarinic acid (RA) is one of toxicity, they may also initiate and/or amplify inflammation
its main phenolic compounds [3]. via the up-regulation of several genes involved in the inflam-
Two studies have evaluated the kinetics of rosmarinic matory response [8].
acid when administered orally to rats [4,5]. These studies A vast amount of circumstantial evidence implicates oxy-
showed that rosmarinic acid was readily absorbed in the gen-derived free radicals and high-energy oxidants as media-
gastrointestinal tract (according to Konishi and Kobayashi tors of inflammation, shock and ischaemia/reperfusion injury
[6], it crosses intestinal epithelium by passive diffusion) and [9]. Antioxidant effects of extracts of R. officinalis have been
reaches the peak plasma concentration at 0.5 hr post-admin- reported in in vitro studies, exhibiting scavenging of superox-
istration. Metabolites formed are a result of glucuronidation, ide anion, peroxynitrite and nitric oxide [10–13].
sulphation and methylation of rosmarinic acid and are then Also, reactive oxygen species play a major role in the organ
eliminated in the urine. The effect of R. officinalis and ros- injury of many inflammatory-mediated events, including sys-
marinic acid on metabolizing enzymes was also studied in temic inflammation and ischaemia–reperfusion [9], particularly
Wistar rats [7]. This study demonstrated that the extract of of the liver [14]. Hepatic ischaemia–reperfusion is a major
R. officinalis was able to induce the enzymes CYP1A1, component of injury in vascular occlusion both during liver
CYP2B1/2, CYP2E1, glutathione S-transferase and UDP-glu- surgery and during liver transplantation. The pathophysiology
curonosyl transferase, but this effect was not observed of hepatic ischaemia–reperfusion includes several mechanisms
when rosmarinic acid was administered alone. The authors that contribute to various degrees of the overall organ damage,
have attributed this effect to the presence of flavones and and formation of reactive oxygen species (ROS) and oxidant
monoterpenes. stress are the most invoked disease mechanisms in ischaemia–
reperfusion injury [15].
Generation of reactive oxygen species during the reperfu-
Author for correspondence: Bruno Sepodes, Faculty of Pharmacy,
University of Lisbon, Avenida Prof. Gama Pinto, 1649-003 Lisbon, sion process can be prevented or attenuated, and that has been
Portugal (fax +351 217946470, e-mail bsepodes@ff.ul.pt). shown to reduce liver injury in experimental models of ischae-

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
EFFECT OF ROSMARINIC ACID IN INFLAMMATION 399

mia–reperfusion [9,16]. Therefore, given the potential antioxi- detector (Shimadzu, RF-535 Kyoto, Japan) and an electrochemical
dant and anti-inflammatory effects of R. officinalis and ros- detector (Dionex, ED40 Bannockburn IL, USA) with a vitreous
carbon electrode were used. Separations were performed at 35°C
marinic acid, they might have protective effects in the injury
with a LiChrospher C18 (5 lm, 250 mm 9 4 mm i.d.) column from
sustained by the liver subjected to ischaemia–reperfusion. Merck Darmstadt, Germany with a guard column of the same type.
Lungs represent the first frontier between oxygen entry into The samples were injected using a 20-lL loop. The separations
the organism and delivery to the mitochondria and are there- were carried out with a flow rate of 700 lL/min., and the mobile
fore exposed to higher concentrations of oxygen and reactive phase consisted of a gradient mixture of eluent A (phosphoric acid
oxygen species [17], leading to the amplification of inflamma- 0.1%) and eluent B (phosphoric acid–acetonitrile–water 5:400:595,
v/v/v). The following gradient of eluents was used: 0–15 min. from
tory processes [18]. A disruption of oxidative balance was
0 until 20% eluent B; 10 min. with 20% eluent B; 25–70 min. from
found to be important in the pathogenesis of lung inflamma- 20 until 70% eluent B; 70–75 min. with 70% eluent B; 75–85 min.
tory diseases, such as acute lung injury (ALI) and acute respi- from 70 until 100% eluent B; 85–90 min. with 100% eluent B.
ratory distress syndrome (ARDS) [19], pneumonia, burn, Diode array detection (DAD) was performed between 200 and
chronic obstructive pulmonary disease and lung ischaemia– 800 nm. Electrochemical detection was programmed for a linear
reperfusion [20–22]. variation of 1.0 V at 1.0 V in 1.00 sec. (detection by integrated
voltammetry using a cyclic variation of the potential). The data
Our aim was to determine the in vitro antioxidant activity
acquisition systems were ChromQuest version 4.0 (Thermo Finnigan
of a R. officinalis extract and rosmarinic acid and to evaluate Surveyor) for the diode array detector and software 4880 (Unicam
and compare their in vivo anti-inflammatory effect on a local Linda-a-Velha, Portugal) for the electrochemical and fluorescence
model of inflammation – the carrageenin-induced paw oedema detectors. A standard solution of rosmarinic acid was prepared
in the rat. We further evaluated the protective effect of ros- (180 p.p.m.), and the peaks of both the standard solution and
marinic acid administration in a hepatic ischaemia–reperfusion sample solution (diluted 1:40) were compared.
model and in a thermal injury model in the rat, taking into
Liquid chromatography with mass spectrometry (LC-MS). A Waters
account the previously described role of oxidative stress in
HPLC equipment (Waters 2695) with a DAD detector (Waters 2996
these systemic inflammatory injuries. Milford, USA) and a Micromass MS equipment (Micromass,
Quattro micro API) were used. Separations were performed with a
Waters dC18 column (5 lm, 2.1 9 150 mm). The samples were
Materials and Methods injected using a loop of 20 lL. The separations were carried out
Chemicals. Rosmarinic acid was obtained from Sigma-Aldrich with a flow rate of 700 lL/min., and the mobile phase consisted of
Quımica SA (Sintra, Portugal). Dried leaves of R. officinalis were a gradient mixture of eluent A (formic acid 0.5%) and eluent B
obtained from Ervital – Plantas Aromaticas e Medicinais, Lda. (Mezio, (acetonitrile). The following gradient of eluents was used: 0–
Portugal). Apyrogenic saline (0.9% NaCl) was obtained from B. Braun 60 min. from 5 until 20% eluent B; 60–80 min. from 20 to 28%
Medical Lda. (Queluz, Portugal). Unless otherwise stated, all other eluent B; 80–82 min. from 28 until 30% eluent B; 82–87 min. with
compounds were obtained from Sigma-Aldrich Quımica SA (fig. 1). 30% eluent B; 87–95 min. from 30 until 5% eluent B; 95–
100 min. with 0% eluent B. Diode detection was performed
between 210 and 600 nm.
Production of the R. officinalis extract. A methanolic extract was
prepared with the dried leaves of R. officinalis and performed with a
continuous Soxhlet extractor equipment (Soxtherm Automatic; Characterization of the antioxidant capacity.
Gerhardt Bonn, Germany). The extraction was performed boiling 7 g
Total phenolic content. The total concentrations of phenolic
of R. officinalis dried leaves, with 50 mL of methanol resulting in an
compounds present in the natural extracts were determined according
extract of 20 mL.
to the Folin–Ciocalteu colorimetric method [23]. Briefly, the
appropriate diluted solutions of extracts were oxidized with a Folin–
Characterization of the chemical profile of the R. officinalis extract. Ciocalteu reagent (Panreac, Barcelona Spain), and the reaction was
neutralized with sodium carbonate. The absorbance of the samples
High-performance liquid chromatography (HPLC-UV). Acetonitrile was measured at 765 nm on a spectrophotometer (GenesysTM 10UV
and methanol were acquired from Lab-Scan (Dublin, Ireland), and ThermoScientific, Waltham MA, USA) after 30 min. at 40°C. Gallic
phosphoric acid was acquired from Riedel-deHaen (Seelze, acid (Fluka, Seelze, Germany) was used as standard, and the result
Germany). Deionized water (conductivity of 0.050) was obtained by was expressed as means of three replicates (mg of gallic acid equiv/L
a Milli-Q system (Millipore, Molsheim, France). A Surveyor of extract – mg GAE/L) [20].
equipment from Thermo Finnigan with a diode array detector
(Thermo Finnigan Surveyor, San Jose, CA, USA), a fluorescence
Oxygen radical absorbance capacity (ORAC). ORAC assay was used
to evaluate the antioxidant capacity of the samples towards peroxyl
radicals. The assay was carried out using the method described by
Serra et al. [24], which measures the ability of the antioxidant species
present in the sample to inhibit the oxidation of FL catalysed by
AAPH-generated peroxyl radicals (ROO˙). The composition of the
reaction mixture was 6.3 9 10 8 M FL, 1.28 9 10 2 M AAPH
(prepared in 75 mM PBS, pH 7.4) and the appropriate diluted sample,
making up a total volume of 1.8 mL. The reaction was started by the
addition of AAPH to the mixture placed in a 10-mm-wide
fluorescence cuvette at 37°C. Fluorescence emitted by the reduced
form of FL was measured and recorded every 1 min. at the emission
Fig. 1. Chemical structure of rosmarinic acid. wavelength of 515 nm and excitation wavelength of 493 nm

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
400 JOAO ROCHA ET AL.

(fluorescence spectrophotometer with thermostatic bath, model Cary Carrageenin-induced paw oedema. The carrageenin-induced paw
Eclipse; Varian Santa Clara CA, USA) for a period of 30 min. oedema of the rat hind paw is a suitable model to study acute local
Phosphate buffer (PBS) was used as a blank, and 1, 5, 12.5, 25 and inflammation and widely considered to be one of the most useful
50 lM Trolox solutions were used as control standards. All samples, models in the evaluation of anti-inflammatory activity of
including the blank and the controls, were analysed in triplicate. Final investigational compounds [30,31].
ORAC values were calculated using a regression equation between the Paw oedema was induced by a single subplantar injection into the
Trolox concentration and the net area under the FL decay curve. Data rat left hind paw of 0.1 mL of a 1% k-carrageenin sterile saline solu-
are expressed as millimolar of Trolox equivalent antioxidant capacity tion. Paw volume was measured by means of a volume displacement
(TEAC), and the experiment was performed in triplicate [24]. method using a plethysmometer (Digital Plethysmometer LE7500; Le-
tica Scientific Instruments (Reagente 5, Porto, Portugal)). Paw volume
Hydroxyl radical absorbance capacity (HORAC). The HORAC assay was measured immediately after the injection of carrageenin (V0 or
was based on a previously reported method [24,25], modified for the basal volume) and 6 hr later (V6 hr).
FL800 microplate fluorescence reader (Bio-Tek Instruments, Isaza, Paw oedema was expressed as percentage of increase in paw vol-
Portugal). The FL800 microplate fluorescence reader was used with ume 6 hr after carrageenin injection relative to the basal values
fluorescence filters for an excitation wavelength of 485  20 nm and according to the equation: % = (V6 hr V0)/V0 9 100.
an emission wavelength of 530  25 nm, and the plate reader was The rats were randomly allocated into eight experimental groups as
controlled by software Gen5 (Biotek, Winnoski VT, USA. Caffeic described:
acid was used as the standard as it provides a wider linear range as 1 Control Group: animals were subjected to subplantar injection into
compared to gallic acid. Data were expressed as mM of caffeic acid the rat left hind paw of 0.1 mL sterile saline and administered
equivalents (CAE). All samples were analysed as triplicates. with vehicle dimethyl sulfoxide – DMSO 10% (1 mL/kg, p.o.)
(n = 6);
Neutrophil oxidative burst. Isolation of human neutrophils. 2 Carrageenin Group: animals subjected to paw oedema induction
Following informed consent, venous blood was collected from healthy by injection into the rat left hind paw of 0.1 mL of k-carrageenin
volunteers by antecubital venipuncture, into K3EDTA vacuum tubes. (1%) and administered with vehicle (1 mL/kg, p.o.) (n = 10);
The isolation of human neutrophils was performed by the gradient 3 RA10 Group: animals subjected to paw oedema induction and
density, as previously reported [26,27]. pre-treated with rosmarinic acid (10 mg/kg, p.o.) 30 min. before
k-carrageenin injection (n = 8);
Evaluation of neutrophils’ oxidative burst. The chemiluminescent 4 RA25 Group: animals subjected to paw oedema induction and
probe luminol has been thoroughly studied and used for monitoring the pre-treated with rosmarinic acid (25 mg/kg, p.o.) 30 min. before
production of reactive species by neutrophils, namely the superoxide k-carrageenin injection (n = 8);
anion radical (O2˙ ), hydrogen peroxide (H2O2), hydroxyl radical 5 RA50 Group: animals subjected to paw oedema induction and
(HO˙), hypochlorous acid (HOCl), nitric oxide (˙NO) and peroxynitrite pre-treated with rosmarinic acid (50 mg/kg, p.o.) 30 min. before
anion (ONOO ) [28]. The measurement of neutrophils’ oxidative burst k-carrageenin injection (n = 8);
was undertaken by chemiluminescence, by monitoring ROS-induced 6 E10 Group: animals subjected to paw oedema induction and pre-
oxidation of luminol, according to a previously described procedure treated with R. officinalis extract (10 mg/kg, p.o.) 30 min. before
[29]. The reaction mixtures contained neutrophils (1 9 106 cells/mL) k-carrageenin injection (n = 8);
and the following reagents at the indicated final concentrations (in a
7 E50 Group: animals subjected to paw oedema induction and pre-
final volume of 250 lL): tested compounds at various concentrations, treated with R. officinalis extract (50 mg/kg, p.o.) 30 min. before
luminol (500 lM) and phorbol myristate acetate (PMA) (160 nM).
k-carrageenin injection (n = 8);
Cells were pre-incubated with luminol and the tested compounds for
8 Indomethacin Group: animals subjected to paw oedema induction
5 min. before the addition of PMA, and the measurements were carried
and pre-treated with indomethacin (10 mg/kg, p.o.) 30 min. before
out at 37°C, under continuous soft shaking. Kinetic readings were
k-carrageenin injection (n = 8).
initiated immediately after cell stimulation. Measurements were taken at
the peak of the curve. This peak was observed at around 10 min. Effects
are expressed as the per cent inhibition of luminol oxidation. Each study Hepatic ischaemia–reperfusion. The rats were randomly allocated into
corresponds to, at least, four individual experiments, performed in five experimental groups as described:
triplicate in each experiment. 1 Control Group: the data were obtained from non-manipulated ani-
mals, that is rats that were not subjected to any surgical procedure
In vivo anti-inflammatory activity. (n = 6).
2 Sham Group: rats that were subjected to the surgical procedures
Experimental methods. Experiments were conducted according to the described below except for liver I/R. Rats were administered vehi-
Home Office Guidance in the Operation of Animals (Scientific cle (saline with 10% DMSO, 1 mL/kg i.v.) 30 min. prior to liver
Procedures) Act 1986, published by Her Majesty’s Stationary Office, I/R (n = 4);
London, UK, and the Institutional Animal Research Committee Guide 3 I/R Group: rats that were subjected to the surgical procedures
for the Care and Use of Laboratory Animals published by the US described below and underwent liver ischaemia for 45 min. fol-
National Institutes of Health (NIH Publication No. 85-23, revised lowed by reperfusion for 2 hr (n = 8);
1996), as well as to the currently adopted EC regulations. Finally, the 4 RA + I/R Group: rats that were administered with RA (25 mg/kg
studies are in compliance with the ARRIVE Guidelines for Reporting i.v.) 30 min. prior to liver I/R (n = 8);
Animal Research summarized at www.nc3rs.org.uk. Paw oedema
5 RA Sham Groups: rats that were administered with (25 mg/kg
studies were carried out using male Wistar rats weighing 100–150 g
i.v.) and that were subjected to the surgical procedures described
(Harlan Iberica, Barcelona, Spain). Hepatic ischaemia–reperfusion
below except for liver I/R (n = 4).
studies were carried out using male Wistar rats weighing 250–300 g
(Harlan Iberica). Thermal injury studies were carried out using male Hepatic ischaemia–reperfusion model was performed as previously
Wistar rats weighing 320–380 g. All animals received a standard diet described [16,32]. Briefly, anaesthetized rats were placed onto a ther-
and water ad libitum. mostatically controlled heating mat (Harvard Apparatus Ltd, Kent,

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
EFFECT OF ROSMARINIC ACID IN INFLAMMATION 401

UK), and body temperature maintained at 37  0.5°C by means of a groups except that room temperature water was used instead of heated
rectal probe attached to a homoeothermic blanket. A tracheotomy was water. After-burn animals were dried and placed over the heating mat for
performed to maintain airway patency and to facilitate spontaneous 6 hr. For these experiments, all rats were anaesthetized with sodium pen-
respiration. The jugular vein was cannulated (PP25, internal diameter tobarbital (60 mg/kg i.p., Eutasil; Sanofi Veterinaria), and anaesthesia
0.40 mm; Portex, Hythe, Kent, UK) for the administration of saline or was maintained by supplementary boli. At the end of all experiments,
anaesthesia as required. A midline laparotomy was performed to care- rats were killed by an overdose of anaesthetic.
fully expose the liver. For the liver I/R surgical procedures, ligament At the of the experiment, blood was collected from the catheter
attachments connecting the liver, diaphragm, abdominal wall and placed in the right carotid artery into a serum SST gel and clot activa-
neighbouring organs were divided. The liver hilus was exposed to find tor tube and was centrifuged (3000 r.p.m. for 10 min. at 4°C) to sepa-
the common hepatic artery and the portal vein. A vascular microclamp rate the serum. The serum was analysed within 24 hr, and liver injury
was used to interrupt the portal venous and arterial hepatic blood sup- was assessed by measurement of the rise in the serum levels of alanine
ply to the three cephalic lobes of the liver during 45 min. The three aminotransferase (ALT, a specific marker for hepatic parenchymal
caudal lobes retained an intact portal inflow and venous out-flow, pre- injury), aspartate aminotransferase (AST, a non-specific marker for
venting the intestinal venous congestion and possible leakage of bacte- hepatic injury) and lactate dehydrogenase (LDH, a marker of non-spe-
ria or bacterial products to the circulation. Reperfusion commenced cific cellular injury), and kidney injury was assessed by measurement
once the vascular clip was removed, and was allowed to proceed for of the rise in the serum levels of creatinine and urea. Measurements of
2 hr. Occlusion was verified visually by the change in the colour of the serum levels of pro-inflammatory cytokines (TNF-a, IL-1b, IL-6)
the liver to a paler shade, and reperfusion by a blush. Other rats were were also taken by ELISAs (Quantikineâ Rat TNF-a/TNFSF1A
subjected to sham operation (sham operated), which underwent surgi- Immunoassay, Quantikineâ Rat IL-1b/IL-1F2 Immunoassay,
cal procedures similar to the I/R group rats but did not undergo I/R of Quantikineâ IL-6 Immunoassay from R&D Systems Inc., Citomed,
the liver clamping and were maintained under anaesthesia for the dur- Lda, Lisboa, Portugal).
ing the experiment. For these experiments, all rats were anaesthetized
with sodium pentobarbital (60 mg/kg i.p., Eutasil; Sanofi Veterinaria Assessment of lung injury. At the end of the experiments,
Porto Salvo, Portugal), and anaesthesia was maintained by supplemen- bronchoalveolar lavage fluid was collected into 15-mL Falcon tubes.
tary i.p. boli. At the end of all experiments, rats were killed by an Collection was performed by instillation of 10 mL of saline at 4°C
overdose of anaesthetic. through the trachea cannula. The samples were centrifuged (3000
At the end of the reperfusion period for the liver I/R experiments r.p.m., 10 min., 4°C), the supernatant was divided into aliquots to
and whenever appropriate regarding other experiments, blood was col- determine albumin and cytokine levels (TNF-a, IL-1b, IL-6), and the
lected from the catheter placed in the right carotid artery into a serum sediment was reconstituted in saline for the polymorphonuclear cell
SST gel and clot activator tube (Becton, Dickinson and Company count. Albumin levels were measured in a Cobas c111 benchtop
Franklin Lakes NJ, USA) and was centrifuged (2000 g for 10 min. at analyzer (Roche Sistemas de Diagnosticos, Lda.). Polymorphonuclear
4°C) to separate the serum. The serum was analysed within 24 hr (Co- cell count was performed on an Automated Hematology System
bas c111 benchtop analyzer; Roche Sistemas de Diagnosticos, Lda. (ADVIA 2120; Siemens Munich, Germany) following May–
Amadora, Portugal), and liver injury was assessed by measuring the Grunwald–Giemsa coloration.
rise in the serum levels of alanine aminotransferase (ALT, a specific Lung tissue samples were obtained at the end of the 6-hr period
marker for hepatic parenchymal injury), aspartate aminotransferase after thermal injury and were fixed in 4% paraformaldehyde in PBS
(AST, a non-specific marker for hepatic injury) and lactate dehydroge- for 72 hr at room temperature, dehydrated through a graded ethanol
nase (LDH, a marker of non-specific cellular injury). series and embedded in paraffin (n = 3 per group). Haematoxylin and
eosin (H&E) staining was performed as previously described [33], and
Thermal injury. The rats were randomly allocated into 4 experimental images were acquired using a brightfield Axioskop microscope (Zeiss,
groups as described: G€ottingen, Germany).
1 Sham Group: rats that were subjected to the surgical procedures For Western blot analysis of Akt and glycogen synthase kinase 3-
described below except for thermal injury. Rats were administered beta (GSK-3b) expression, frozen tissue sample cells were lysed in
vehicle (saline with 10% DMSO, 1 mL/kg i.v.) 5 min. prior to RIPA buffer containing Tris 50 mM (pH 8.0), 5 mM EDTA (pH
thermal injury (n = 9); 8.0), 150 mM NaCl, 1% NP-40, 10% glycerol and 0.1% SDS, and
2 Thermal Injury Group: rats that were subjected to the surgical pro- sonicated for 20 sec. The lysate was centrifuged at 14,000 9 g for
cedures described below and underwent thermal injury (n = 14); 10 min. at 4°C, and the supernatants were collected and stored at
3 RA+Thermal Injury Group: rats that were administered with RA 80°C. Protein concentrations were determined using Nanodrop ND-
(25 mg/kg i.v.) 5 min. prior to thermal injury (n = 14); 1000 (ThermoScientific, Wilmington DE, USA. Cell extracts contain-
ing equal amounts of protein (100–150 lg) were separated on
4 RA Sham Group: rats that were administered with (25 mg/kg i.v.)
sodium dodecyl sulphate–polyacrylamide gel electrophoresis and
and that were subjected to the surgical procedures described below
transferred to a nitrocellulose membrane. The membranes were
except for thermal injury (n = 6).
blocked with 5% non-fat milk, incubated with the primary antibody
Briefly, anaesthetized rats were placed onto a thermostatically con- overnight at 4°C rabbit anti-p-Akt [1:1000 (#12178; Cell Signalling,
trolled heating mat (Harvard Apparatus Ltd), and body temperature Beverly, MA, USA)], rabbit anti-Akt [(1:1000) (#4691; Cell Signal-
maintained at 37  0.5°C by means of a rectal probe attached to a ho- ling)], goat anti-p-GSK-3b [(1:200) (#sc-11757; Santa Cruz, CA,
moeothermic blanket. A tracheotomy was performed to maintain airway USA)], mouse anti-GSK-3b [(1:1000) (#9832; Cell Signalling)] and
patency, facilitate spontaneous respiration and removal of secretions. then with a horseradish peroxidase-labelled secondary antibody for
The jugular vein was cannulated for the administration of saline or anaes- 1 hr at room temperature. After extensive washes, immunoreactive
thesia as required, and the carotid artery was cannulated (PP50, I.D. bands were detected by LumiGLOâ (Cell Signalling) and visualized
0.58 mm; Portex Kent, UK) for haemodynamic monitoring by a pressure by autoradiography with Hyperfilm ECL. Phosphorylation levels of
transducer (MLT0380 BP Transducer; AD Instruments Oxford, UK) and Akt and GSK-3b were analysed by the ratio of the phosphorylated
blood collection at the end of the experiment. A 30% third-degree skin form to total enzyme levels and expressed as fold change.
burn was induced by immersing dorsal-shaved skin into 99°C water for
10 sec. using a synthetic foam template after surgical procedure. The Evaluation of factor nuclear kappa B (NF-kB) activity. Nuclear
sham control groups were submitted to identical procedures as the other extracts were prepared according to the rapid Dignam method [34].

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
402 JOAO ROCHA ET AL.

Briefly, at the end of the incubation, cells were resuspended in under non-reducing conditions. After electrophoresis, gels were
400 lL ice-cold buffer [10 mM HEPES (pH 7.9), 10 mM KCl, washed for 1 hr with 2.5% Triton X-100 (in 50 mM Tris pH 7.4;
0.1 mM EDTA, 0.1 mM EGTA, 1 mM dithiothreitol (DTT) and 5 mM CaCl2; 1 lM ZnCl2) to remove SDS and renature the matrix
0.5 mM phenylmethylsulphonyl fluoride (PMSF)] for 20 min. on ice metalloproteinases (MMP) species in the gel. Then, the gels were
and vortexed vigorously for 5 sec. after the addition of 25 lL Nonidet incubated in the developing buffer (50 mM Tris pH 7.4; 5 mM
P40 (10%). Cell lysates were centrifuged at 10,000 9 g for 30 sec. at CaCl2; 1 lM ZnCl2) overnight to induce gelatine lysis. For enzyme
4°C, and the pelleted nuclei were lysed with 50 lL ice-cold buffer activity analysis, the gels were stained with 0.5% Coomassie
[10 mM HEPES (pH 7.9), 0.4 M NaCl, 10 mM EDTA, 10 mM Brilliant Blue R-250 (Sigma-Aldrich Quimica SA, Sintra Portugal)
EGTA, 1 mM DTT, 1 mM PMSF] for 20 min. at 4°C. Nuclear and destained in 30% ethanol/10% acetic acid/H2O. Gelatinase
lysates were centrifuged at 14,000 9 g for 10 min. at 4°C, and the activity, detected as a white band on a blue background, was
nuclear proteins recovered in the supernatants were stored at 80°C. quantified by computerized image analysis and normalized with total
After protein determination using the protein assay kit (Bio-Rad, cellular protein. MMP-9 was detected as the band with 92 kDa.
Hercules, CA, USA), the p65 NF-jB subunit expression in each
compartment was analysed by Western blot as previously described Statistical analysis. The results were expressed as the mean  S.E.M.
[35]. Extracts containing equal amounts of protein (50 lg) were and were compared using a one-factorial ANOVA test, followed by a
separated on sodium dodecyl sulphate–polyacrylamide gel Bonferroni’s post hoc test performed with a GraphPad Prism
electrophoresis and transferred to a nitrocellulose membrane. The Statistical Package (version 5.0 GraphPad Software, La Jolla CA,
membranes were blocked with 5% non-fat milk, incubated with the USA)). A p-value less than 0.05 was considered to be statistically
primary antibody overnight at 4°C [rabbit anti-p65 NF-jB subunit significant.
(1:1000) (SC-372; SantaCruz Biotechology)] and then with a
horseradish peroxidase-labelled secondary antibody for 1 hr at room
temperature. After extensive washes, immunoreactive bands were Results
detected by LumiGLOâ (Cell Signalling) and visualized by
autoradiography with Hyperfilm ECL. Results were normalized to Characterization of the chemical profile of the R. officinalis
total protein lane content measured following Amido Black staining. extract.
Chemical characterization of the extract by HPLC-UV and
Evaluation of cell death. Cell death was evaluated by caspase-3 HPLC-MS revealed a high concentration of rosmarinic acid
activation as previously described [36]. The activity of caspase-3 was (4684 p.p.m., corresponding to 1.34 g of rosmarinic acid per
determined in tissue homogenates by enzymatic cleavage of
100 g dried leaves) as demonstrated by the chromatographic
chromophore p-nitroanilide (pNA) from the substrate Ac-DEVD-pNA
for caspase-3, according to manufacturer’s instructions. The proteolytic profile of a 280-nm HPL-UV (fig. 2), indicating that the
reaction was carried out in protease assay buffer [50 mM HEPES (pH extraction procedure had optimal conditions for the preferen-
7.4); 100 mM NaCl; 0.1% (w/v) CHAPS; 10 mM DTT; 0.1 mM tial extraction of rosmarinic acid. Chemical profiling of the
EDTA; 10% (v/v) glycerol], containing 2 mM specific substrate. After extract also revealed the presence of other phenolic com-
the incubation of the reaction mixtures for 1–2 hr at 37°C, the formation
pounds, although in minor concentrations (caffeic acid, p-
of pNA was measured in a microplate reader (PR 2100; BioRad
Laboratories, Inc.) at k=405 nm with a reference filter at 620 nm.
coumaric acid, chlorogenic acid, ellagic acid, ferulic acid,
Readings were normalized to total protein content determined using a rosmanol, rutin, luteolin, methoxyluteolin, nepitin).
protein assay kit (Bio-Rad) according to the manufacturer’s
specification, and expressed as fold change of respective control.
Characterization of the antioxidant capacity.
The antioxidant capacity assays demonstrated that both ros-
Gelatin zymography (Metalloproteinases assay). Activity of matrix
metalloproteinase (MMP-9) was assessed by gelatin zymography as marinic acid and R. officinalis extract have high antioxidant
described before [37]. Aliquots of tissue homogenates were analysed capacities (table 1), although R. officinalis extract presented a
by SDS-PAGE zymography in 0.1% gelatine–10% acrylamide gels much higher activity.

Rosmarinic acid

Fig. 2. Chromatogram profile (1:10) of the Rosmarinus officinalis extract on wavelength 280 nm.

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
EFFECT OF ROSMARINIC ACID IN INFLAMMATION 403

Neutrophil oxidative burst. Hepatic ischaemia–reperfusion.


Pre-incubation of the neutrophils with rosmarinic acid resulted In sham-operated rats, the surgical procedure did not produce
in an inhibition of luminol oxidation, induced by neutrophil a significant change in serum levels of AST, ALT or LDH
activation by phorbol myristate acetate (PMA) (fig. 3). The when compared with the control group. When compared with
reduction was statistically significant compared with control sham-operated rats, I/R of the liver resulted in significant rises
neutrophils (incubation with PMA only) and exhibits a con- in the serum levels of the marker enzymes AST, ALT and
centration–response relation. The inhibitory concentration 50% LDH, demonstrating the development of hepatocellular injury.
(IC50) was 1.2  0.3 lM. In rats subjected to I/R pre-treated with rosmarinic acid, the
serum rise in AST, ALT and LDH was significantly reduced
by 35%, 35% and 45%, respectively. Rosmarinic acid by
Carrageenin-induced paw oedema.
itself, when administered to rats not subjected to I/R injury,
As expected, intraplantar injection of carrageenin in rats led to
had no significant effect on AST, ALT and LDH serum levels
an increase in paw volume after 6 hr (114  11.2%) when
(fig. 6).
compared with the control group. This increase was signifi-
cantly reduced in a dose-dependent manner by pre-treatment
with rosmarinic acid and R. officinalis extract, with the most Thermal injury model.
significant effect observed at the doses 25 and 50 mg/kg
Effect of rosmarinic acid on liver and kidney injury. When
(increase in paw volume of 45.3  12.8% and 44.4  13%,
compared with sham-operated rats, thermal injury resulted in
respectively) (fig. 4).
significant rises in the serum levels of the marker enzymes AST
A comparison of the effect of a single administration of ros-
and ALT, demonstrating the development of hepatocellular
marinic acid (25 mg/kg) with the effect of lycopene (50 mg/
injury. No significant rise was detected in rats subjected to
kg), tempol (30 mg/kg), Trolox (30 mg/kg) and indomethacin
rosmarinic acid treatment in comparison with sham-operated
(10 mg/kg), known antioxidant and anti-inflammatory sub-
rats, indicating that rosmarinic acid administration prevented the
stances, showed that rosmarinic acid administration reduced
development of hepatocellular injury. Termal injury also led to
oedema formation at the same magnitude as those substances
a significant rise in creatinine and urea serum levels indicating
(fig. 5).
the development of kidney injury or dysfunction. In rats
subjected to treatment with rosmarinic acid, the rise in
Table 1.
creatinine serum level was significantly attenuated, although
Characterization of the antioxidant capacity of rosmarinic acid and urea levels were not statistically different from rats subjected
Rosmarinus officinalis extract. only to thermal injury. Rats subjected to thermal injury also
exhibited a rise in serum levels of LDH, a non-specific marker
Total phenolic
content ORAC HORAC of cell injury. No significant rise in LDH levels was detected in
rats subjected to rosmarinic acid treatment in comparison with
Rosmarinic – 112 mM TEAC 66 mM CAE
sham-operated rats (fig. 7).
acid
R. officinalis 62.5 mM GAE 1384 mM TEAC 1516 mM CAE
extract Effect of rosmarinic acid on cytokine serum levels. When
compared with sham-operated rats, thermal injury resulted in
GAE, gallic acid equivalents; CAE, caffeic acid equivalents; TEAC,
Trolox equivalent antioxidant capacity. significant rises in the serum levels of pro-inflammatory

100
Luminol oxidation by activated
neutrophils (% inhibition)

***
75 ***
***

50 **

25

0
0.2 0.5 0.9 1.9 3.8 7.5
μM

Fig. 3. Inhibitory effect of rosmarinic acid (0.2–7.5 lM) on human neutrophils’ oxidative burst stimulated with PMA. **p < 0.01 and
***p < 0.001 compared with the control assay (PMA alone). The values are given as the mean  S.E.M. (n ≥ 4).

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
404 JOAO ROCHA ET AL.

Fig. 4. Effect of Rosmarinus officinalis extract and rosmarinic acid administration on the rat paw oedema development elicited by carrageenin 6 hr
after oedema induction. Effect of a single administration of R. officinalis extract (E10 mg/kg, n = 8; and E25 mg/kg, n = 8, p.o.) in comparison
with the effect of a single administration of rosmarinic acid (RA10 mg/kg, n = 8; RA25 mg/kg, n = 8; RA50 mg/kg, n = 8, p.o.) and indometha-
cin (10 mg/kg, p.o., n = 8). The data are presented as means with their standard errors. *p < 0.001 versus control group; ‡p < 0.01 versus control
group; #p < 0.01 versus carrageenin group.

130 *
120
110
100
% paw volume increase

90
80
70
# #
60 # #
#
50
40
30
20
10
0
–10 Control Carrageenan RA 25 Lycopene Tempol Trolox Indomethacin

Fig. 5. Comparison of the anti-inflammatory effect of rosmarinic acid with known antioxidant and anti-inflammatory substances. Effect of a single
administration of rosmarinic acid (RA25 mg/kg, n = 8; p.o.), lycopene (50 mg/kg, n = 8; p.o.), tempol (30 mg/kg, n = 8; p.o.), Trolox (30 mg/kg,
n = 8; p.o.) and indomethacin (10 mg/kg, p.o., n = 8). The data are presented as means with their standard errors. *p < 0.001 versus control group;
#
p < 0.01 versus carrageenin group (Bignotto et al., 2009).

cytokines (TNF-a, Il-1b and IL-6). No significant rise was detected in rats subjected to rosmarinic acid treatment in
detected in rats subjected to rosmarinic acid treatment in comparison with sham-operated rats (fig. 10).
comparison with sham-operated rats (fig. 8).
Effect of rosmarinic acid on the polymorphonuclear (PMN)
Effect of rosmarinic acid on cytokine bronchoalveolar lavage cell presence on bronchoalveolar lavage fluid (BALF). When
fluid (BALF) levels. When compared with sham-operated compared with sham-operated rats, thermal injury resulted in
rats, thermal injury resulted in significant rises in the BALF significant rises in the leucocyte and neutrophil number in
levels of pro-inflammatory cytokines (TNF-a, IL-1b and IL- BALF and also an increase in neutrophil percentage. No
6). No significant rise in the BALF levels of TNF-a and significant rise was detected in these parameters in rats
IL-6 was detected in rats subjected to rosmarinic acid subjected to rosmarinic acid treatment in comparison with
treatment in comparison with sham-operated rats, and the sham-operated rats (fig. 11).
rise in the BALF levels of IL-1b was significantly reduced
(fig. 9). Effect of rosmarinic acid on lung histology. When compared
with sham-operated rats, thermal injury resulted in structural
Effect of rosmarinic acid on albumin concentration on changes that indicate lung tissue injury, namely neutrophil
bronchoalveolar lavage fluid (BALF). When compared with accumulation on the peri-alveolar zone, hyaline membrane
sham-operated rats, thermal injury resulted in significant rises formation with thickening of the alveolar wall and intra-
in the BALF levels of albumin demonstrating the disruption alveolar (clots) and tissue haemorrhage (patchy formation). In
of the alveolo-capillary membrane. No significant rise was rats subjected to rosmarinic acid treatment, a marked reduction

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
EFFECT OF ROSMARINIC ACID IN INFLAMMATION 405

2500
2500
*
2000 *
2000
* #
AST (U/L)

1500

ALT (U/L)
* # 1500

1000
1000

500 500

0 0
Control Sham I/R RA + I/R RA Control Sham I/R RA + I/R RA

9000
*

7500

6000 * #
LDH (U/L)

4500

3000

1500

0
Control Sham I/R RA + I/R RA

Fig. 6. Effect of rosmarinic acid administration (25 mg/kg, i.v.) on the serum levels of aspartate aminotransferase (AST), alanine aminotransferase
(ALT) and lactate dehydrogenase (LDH), in the rat model of hepatic ischaemia–reperfusion. Control group, n = 6; sham group, n = 4; I/R group,
n = 8; RA+I/R group, n = 8; RA group, n = 4. *p < 0.001 versus sham group; #p < 0.01 versus I/R group.

of these changes was observed. Haemorrhage areas in patches acid, there was a significant increase in the expression and
are still observable but occupy a lesser area and are more rare. nuclear translocation of NF-jB (fig. 15).
Neutrophil accumulation on the peri-alveolar zone was also
rare, and no alveoli were found with thickening of the Effect of rosmarinic acid on the activation of caspase-
alveolar wall (fig. 12). 3. When compared with sham-operated rats, thermal injury
resulted in a significant increase in the activation of caspase-3.
Effect of rosmarinic acid on the activation of Akt. When In rats subjected to treatment with rosmarinic acid, although a
compared with sham-operated rats, thermal injury resulted in a trend appears to exist, there was no significant decrease in the
significant rise in the activation of Akt (expressed as increased activation of caspase-3 (fig. 16).
ratio of p-Akt/Akt). In rats subjected to treatment with
rosmarinic acid, the rise in Akt activation was significantly Effect of rosmarinic acid on the activation of
attenuated (fig. 13). metalloproteinase-9 (MMP-9). When compared with sham-
operated rats, thermal injury resulted in significant rises in the
Effect of rosmarinic acid on the activation of GSK-3b. When activation of MMP-9. No significant rise was detected in rats
compared with sham-operated rats, thermal injury resulted in a subjected to rosmarinic acid treatment in comparison with
significant decrease in the activation of GSK-3b (expressed as sham-operated rats (fig. 17).
increased ratio of p-GSK-3b/GSK-3b). In rats subjected to
treatment with rosmarinic acid, there was no significant
Discussion
change in the activation of GSK-3b (fig. 14).
Inflammatory response develops along several stages,
Effect of rosmarinic acid on the activation of NF-jB. When apparently mediated through different mechanisms. The
compared with sham-operated rats, thermal injury resulted in a carrageenin-induced paw oedema model induces a biphasic
significant increase in the expression and activation of NF-jB oedema consisting of an early-phase (up to 2 hr) followed by
(expressed as increased nuclear translocation of the p65 a more sustained late-phase (2–6 hr). The late-phase is related
NF-jB subunit). In rats subjected to treatment with rosmarinic to neutrophil infiltration [38] and production of reactive

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
406 JOAO ROCHA ET AL.

2500
2500
*
2000 *
2000
* #
1500
AST (U/L)

1500

ALT (U/L)
* #

1000
1000

500 500

0 0
Control Sham I/R RA + I/R RA Control Sham I/R RA + I/R RA

9000
*

7500

6000 * #
LDH (U/L)

4500

3000

1500

0
Control Sham I/R RA + I/R RA

Fig. 7. Effect of rosmarinic acid administration (25 mg/kg, i.v.) on the serum levels of aspartate aminotransferase (AST), alanine aminotransferase
(ALT), lactate dehydrogenase (LDH), creatinine and urea, in the rat model of thermal injury. Sham group, n = 9; thermal injury (TI) group,
n = 14; RA + TI group, n = 14; RA group, n = 6. *p < 0.05 versus sham group; #p < 0.05 versus TI group.

250 175 *
*
150
200
125
TNF-α (pg/mL)
IL-6 (pg/mL)

150
100 #
#

100 75

50
50
25

0 0
Sham TI RA + TI Sham TI RA + TI

500 *

400
IL-1β (pg/mL)

300 #

200

100

0
Sham TI RA + TI

Fig. 8. Effect of rosmarinic acid administration (25 mg/kg, i.v.) on the serum levels of TNF-a, IL-1b and IL-6, in the rat model of thermal injury. Sham
group, n = 9; thermal injury (TI) group, n = 14; RA + TI group, n = 14; RA group, n = 6. *p < 0.05 versus sham group; #p < 0.05 versus TI group.

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
EFFECT OF ROSMARINIC ACID IN INFLAMMATION 407

250 * 25 *

200 20
# #

TNF-α (pg/mL)
IL-6 (pg/mL)

150 15

100 10

50 5

0 0
Sham Q AcR + Q Sham Q AcR + Q

450 *
400

350

300
IL-1β (pg/mL)

250 #

200

150

100

50

0
Sham Q AcR + Q

Fig. 9. Effect of rosmarinic acid administration (25 mg/kg, i.v.) on the bronchoalveolar lavage fluid levels of TNF-a, IL-1b and IL-6, in the rat
model of thermal injury. Sham group, n = 9; thermal injury (TI) group, n = 14; RA + TI group, n = 14; RA group, n = 6. *p < 0.05 versus sham
group; #p < 0.05 versus TI group.

species such as hydrogen peroxide, superoxide radical, perox- the inflammatory process [41,42] and contribute greatly to tis-
ynitrite [39] and pro-inflammatory prostanoids [40]. This sue injury caused by inflammation [43]. In fact, reactive oxy-
model has been used greatly in research and has become the gen species are already considered as a valid therapeutic target
mainstay in the elucidation of pharmacodynamic properties in in the development of new drugs for the treatment of diseases
the support of non-clinical development of most of the non- with a strong inflammatory component [44,45], and several
steroidal anti-inflammatory drugs [30]. plant-derived products are already in a late stage for drug
Several in vitro and in vivo studies have shown that reactive development aiming for therapeutic indications where oxida-
oxygen species play a central role in the physiopathology of tive stress and inflammation might play a significant role [46].
The high concentration of phenolic acids in extracts of R. offi-
cinalis has been considered to be related to its antioxidant
4
capacity [47,48], anti-inflammatory [12] and antibacterial
Albumin concentration on BALF (mg/dL)

activity [47]. Given the central role of oxidation and reactive


*
oxygen species in inflammation and the known antioxidant
3
effects of rosmarinic acid and R. officinalis, we aimed to eval-
#
uate their effect in this model of acute local inflammation.
#
2
Our results suggest that pre-treatment with rosmarinic acid or
R. officinalis extract significantly reduces the increase in paw
volume by inhibiting the inflammatory processes associated
1
with oedema formation.
Administration of an extract of R. officinalis and rosmarinic
acid led to an anti-inflammatory effect as demonstrated by a
0
60% reduction in paw oedema volume in comparison with
Sham TI RA + TI RA control animals. Administration of the extract, in a dose equiv-
alent to 25 mg/kg of rosmarinic acid, exhibited the same mag-
Fig. 10. Effect of rosmarinic acid administration (25 mg/kg, i.v.) on
nitude of effect, suggesting that it was RA that mainly
the bronchoalveolar lavage fluid levels of albumin, in the rat model of
thermal injury. Sham group, n = 9; thermal injury (TI) group, n = 14; contributed to the anti-inflammatory effect observed. Given
RA + TI group, n = 14; RA group, n = 6. *p < 0.05 versus sham the naturally higher antioxidant capacity of the extract in rela-
group; #p < 0.05 versus TI group. tion to rosmarinic acid alone, it is possible to assume that the

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
408 JOAO ROCHA ET AL.

Fig. 11. Effect of rosmarinic acid administration (25 mg/kg, i.v.) on the bronchoalveolar lavage fluid number and percentage of polymorphonuclear
cells, in the rat model of thermal injury. Sham group, n = 9; thermal injury (TI) group, n = 14; RA + TI group, n = 14; RA group, n = 6.
*p < 0.05 versus sham group; #p < 0.05 versus TI group.

anti-inflammatory effect exhibited in this model of acute has also demonstrated the protective effect of rosmarinic
inflammation might not be solely related to the antioxidant acid on liver injury. Administration of rosmarinic acid
capacity, but also to other mechanisms that may need further (135 mg/kg, p.o.) reduced lipopolysaccharide-induced liver
investigation. The same magnitude of effect exhibited by ros- injury in D-galactosamine-sensitized mice, and the liver pro-
marinic acid compared with other known and extensively stud- tection exhibited by RA was related to the scavenging or
ied antioxidant and anti-inflammatory substances (tempol, reducing activities of superoxide and peroxynitrite [50].
Trolox, lycopene and indomethacin) suggests that rosmarinic Osakabe et al. also observed that the magnitude of rosmari-
acid may have the potential to reduce inflammatory-induced nic effect was similar to that of administration of an extract
tissue injury in other inflammation models and through differ- of Perilla frutescens with an equivalent dose of RA, attrib-
ent mechanisms of action. uting the protective effect of the extract to its main constit-
Most interestingly, rosmarinic acid was also able to inhi- uent, rosmarinic acid.
bit the inflammatory process associated with hepatic ischae- Thermal injury has a very severe clinical prognosis and lim-
mia–reperfusion, thereby reducing liver injury sustained after ited therapeutic options. Severe thermal injury leads frequently
reperfusion. Liver injury caused by ischaemia–reperfusion to systemic inflammatory response syndrome with subsequent
consists of an interruption of blood supply to the liver fol- multi-organ dysfunction syndrome and a potentially fatal out-
lowed by reperfusion. When the blood supply is restored, come, being the main cause of death in burned patients [51].
the organs are usually subjected to a further insult, aggra- Acute lung injury (ALI) and acute respiratory distress
vating the injury created within the ischaemic period [32]. syndrome (ARDS) are at the top of the list of early complica-
This reperfusion insult is a direct consequence of a complex tions in burned patients and associated with a high mortality
interplay between different mechanisms. The initial phase [52–54].
(until 2 hr after reperfusion) is characterized by oxidative Several physiopathological events are considered the main
stress. The destructive effects of ischaemia–reperfusion result contributors in the development of acute lung injury, includ-
from the acute generation of ROS subsequent to reoxygen- ing systemic release of inflammatory mediators (cytokines),
ation. These ROS inflict direct tissue damage and initiate a chemotaxis and activation of neutrophils (oxidative burst)
cascade of deleterious cellular responses leading to inflam- and activation of cellular pathways involved in cell death
mation, cell death and organ failure [49]. A previous work [55].

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
EFFECT OF ROSMARINIC ACID IN INFLAMMATION 409

A B

C D

E F

Fig. 12. Effect of rosmarinic acid administration (25 mg/kg, i.v.) on the lung tissue histological features, in the rat model of thermal injury. (A)
Sham group (1009); (B) sham group (4009); (C) thermal injury group (1009); (D) thermal injury group (4009); (E) rosmarinic acid + thermal
injury group (1009); and (F) rosmarinic acid + thermal injury group (4009). Images are representative of at least 4 experiments performed on dif-
ferent days.

The scalding burn model used in this model is widely cited results revealed inhibition of the neutrophils’ oxidative burst
in the literature [56–58], and we were able to prove that it by rosmarinic acid in a dose-dependent manner suggesting that
leads to the internationally acknowledged main features of an its anti-inflammatory effect might be related, at least in part,
experimental model of ALI/ARDS [59]. by inhibition of neutrophil activation.
We demonstrated that administration of rosmarinic acid was Mechanistic studies performed on the lung tissue in the
able to reduce systemic release of pro-inflammatory cytokines thermal injury model revealed that the beneficial effect
and also attenuated the multi-organ injury (liver, kidney and exhibited by rosmarinic acid might not be related to modu-
lung) induced by scald. Specifically on the lung, the histologi- lation of the PI3K-Akt-GSK-3b pathway but rather through
cal analysis of the lung showed that there was a marked activation of the NF-jB pathway and inhibition of MMP-9
reduction in the histological signs of lung tissue injury with activation. MMP-9 has been shown to regulate the activa-
an appearance closer to sham-operated rats, not subjected to tion of pro-apoptotic factors such as the Fas/FasL complex
thermal injury. [65], and direct inhibition of MMP-9 or its gene transcrip-
Along with the formation of ROS, the infiltration and accu- tion has been shown to be beneficial in several lung injury
mulation of polymorphonuclear leucocytes within the tissues experimental models [66–68]. Given the importance of the
has been considered a hallmark of the acute inflammatory NF-jB pathway on cell survival [69] and its role in the
response, including local inflammation [34] hepatic ischae- crosstalk with several other pathways related to apoptosis,
mia–reperfusion [14,60], acute lung injury [61,62] and sys- cell fate and inflammation [70,71], we might speculate that
temic inflammation [63,64]. A prominent feature of acute rosmarinic acid might exhibit pleiotropic mechanisms and
inflammation is enhanced vascular permeability resulting in act through the modulation of NF-jB closely related path-
oedema formation. Such changes in vascular permeability have ways.
been known to be dependent upon polymorphonuclear leuco- Given that the inflammatory process is a multi-factorial
cyte interactions with the vascular endothelium [63]. Our network of different mediators and that oxidative stress and

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
410 JOAO ROCHA ET AL.

Sham TI RA + TI Sham TI RA + TI
C N C N C N
P-AKT - - 60 kDa
NF-κB- - 65 kDa

AKT total - - 60 kDa


#
20 Cytoplasm
2.5 Nucleus
*

NF-κB protein expression


15
#
pAKT/AKT protein expression

2.0

(fold change)
10
*
(fold change)

1.5

5
1.0

0
0.5 Sham TI RA + TI

Fig. 15. Effect of rosmarinic acid administration (25 mg/kg, i.v.) on


0.0 the lung tissue NF-jB activation, in the rat model of thermal injury.
Sham group, n = 9; thermal injury (TI) group, n = 14; RA + TI
am

TI

TI
+

group, n = 14; RA group, n = 6. *p < 0.05 versus sham group;


Sh

A
R

#
p < 0.05 versus TI group.
Fig. 13. Effect of rosmarinic acid administration (25 mg/kg, i.v.) on
the lung tissue Akt activation, in the rat model of thermal injury.
Sham group, n = 9; thermal injury (TI) group, n = 14; RA + TI
group, n = 14; RA group, n = 6. *p < 0.05 versus sham group;
#
p < 0.05 versus TI group.

Sham TI RA + TI

P-GSK3β - - 46 kDa

GSK3β total - - 46 kDa


Fig. 16. Effect of rosmarinic acid administration (25 mg/kg, i.v.) on
the lung tissue caspase-3 activation, in the rat model of thermal injury.
Sham group, n = 9; thermal injury (TI) group, n = 14; RA + TI
2.5 group, n = 14; RA group, n = 6. *p < 0.05 versus sham group;
#
p < 0.05 versus TI group.
pGSK/GSK protein expression

2.0 #
(fold change)

1.5
neutrophil oxidative burst play an important role in both
models of inflammation used in this work, it is possible to
1.0 speculate that rosmarinic acid might be exhibiting an
anti-inflammatory activity by a net effect, which includes its
0.5 antioxidant properties, inhibition of neutrophil activity, inhi-
bition of MMP-9 activity and modulation of the NF-jB
0.0 pathway.
Therefore, rosmarinic acid administration might be useful in
TI
am

TI

+
Sh

the management of inflammatory processes, and further


A
R

research on anti-inflammatory effects of rosmarinic acid and


Fig. 14. Effect of rosmarinic acid administration (25 mg/kg, i.v.) on
R. officinalis might lead to the discovery of new pharmacolog-
the lung tissue GSK-3b activation, in the rat model of thermal injury.
Sham group, n = 9; thermal injury (TI) group, n = 14; RA + TI ical tools in the treatment of inflammatory diseases. Given the
group, n = 14; RA group, n = 6. *p < 0.05 versus sham group; recognized role of inflammation and oxidative stress in neuro-
#
p < 0.05 versus TI group. degenerative diseases [72], cardiovascular diseases [73],

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
EFFECT OF ROSMARINIC ACID IN INFLAMMATION 411

9 Cuzzocrea S, Riley DP, Caputi AP, Salvemini D. Antioxidant ther-


apy: a new pharmacological approach in shock, inflammation, and
ischemia/reperfusion injury. Pharmacol Rev 2001;53:135–59.
10 Choi HR, Choi JS, Han YN, Bae SJ, Chung HY. Peroxynitrite
scavenging activity of herb extracts. Phytother Res 2002;4:364–7.
11 Nakamura Y, Ohto Y, Murakami A, Ohigashi H. Superoxide scav-
enging activity of rosmarinic acid from Perilla frutescens Britton
Var. acuta f. viridis. J Agric Food Chem 1998;46:4545–50.
12 Peng CH, Su JD, Chyau CC, Sung TY, Ho SS, Peng CC et al.
Supercritical fluid extracts of rosemary leaves exhibit potent anti-
inflammation and anti-tumor effects. Biosci Biotechnol Biochem
2007;71:2223–32.
13 Qiao S, Li W, Tsubouchi R, Haneda M, Murakami K, Takeuchi F
et al. Rosmarinic acid inhibits the formation of reactive oxygen
Fig. 17. Effect of rosmarinic acid administration (25 mg/kg, i.v.) on and nitrogen species in RAW264.7 macrophages. Free Radic Res
the lung tissue metalloproteinase-9 (MMP-9) activation, in the rat 2005;39:995–1003.
model of thermal injury. Sham group, n = 9; thermal injury (TI) 14 Jaeschke H. Mechanisms of liver injury. II. Mechanisms of neutro-
group, n = 14; RA + TI group, n = 14; RA group, n = 6. *p < 0.05 phil-induced liver cell injury during hepatic ischemia-reperfusion
versus sham group; #p < 0.05 versus TI group. and other acute inflammatory conditions. Am J Physiol Gastroin-
test Liver Physiol 2006;6:G1083–8.
15 Jaeschke H, Woolbright BL. Current strategies to minimize hepatic
respiratory diseases [74] and cancer [75], these promising ischemia-reperfusion injury by targeting reactive oxygen species.
results might unveil a potential research field for rosmarinic Transplant Rev (Orlando) 2012;2:103–14.
acid and its clinical applications. 16 Sepodes B, Maio R, Pinto R, Marques C, Mendes-do-Vale J,
McDonald MC et al. Tempol, an intracelullar free radical scaven-
ger, reduces liver injury in hepatic ischemia-reperfusion in the rat.
Acknowledgements
Transplant Proc 2004;36:849–53.
João Rocha was supported by FCT under a Doctoral Grant 17 Ward PA. Oxidative stress: acute and progressive lung injury. Ann
(SFRH/BD/64180/2009). Marisa Freitas acknowledges the N Y Acad Sci 2010;1203:53–9.
financial support by FCT for the Pos-doc grant (SFRH/BPD/ 18 Zimmerman JJ. Oxidant stress in acute lung injury. In: Wong HR,
76909/2011), in the scope of ‘QREN e POPH e Tipologia 4.1 Shanley T (eds). Molecular Biology of Acute Lung Injury, 1st edn.
Springer, Berlin, Germany, 2001;83–99.
e Formaç ão Avanç ada’, cosponsored by FSE and by National
19 Crimi E, Sica V, Slutsky AS, Zhang H, Williams-Ignarro S,
Funds of MCTES. Ignarro LJ et al. Role of oxidative stress in experimental sepsis
References and multisystem organ dysfunction. Free Radic Res
2006;40:665–72.
1 Gonzá lez-Trujano ME, Peña EI, Martínez AL, Moreno J, 20 Bowler RP, Crapo JD. Oxidative stress in allergic respiratory dis-
Guevara-Fefer P, Dé ciga-Campos M et al. Evaluation of the eases. J Allergy Clin Immunol 2002;110:349–56.
antinociceptive effect of Rosmarinus officinalis L. using three dif- 21 Crapo JD. Oxidative stress as an initiator of cytokine release and
ferent experimental models in rodents. J Ethnopharmacol cell damage. Eur Respir J 2003;44:4s–6s.
2007;111:476–82. 22 Rahman I, Kelly F. Biomarkers in breath condensate: a promising
2 Takaki I, Bersani-Amado LE, Vendruscolo A, Sartoretto SM, Diniz new non-invasive technique in free radical research. Free Radic
SP, Bersani-Amado CA et al. Anti-inflammatory and antinocicep- Res 2003;37:1253–66.
tive effects of Rosmarinus officinalis L. essential oil in experimen- 23 Singleton VL, Rossi JA. Colorimetry of total phenolics with phos-
tal animal models. J Med Food 2008;11:741–6. phomolybdic-phosphotungstic acid reagents. Am J Enol Vitic
3 del Ba~ no MJ, Lorente J, Castillo J, Benavente-Garcıa O, del Rıo 1965;16:144–58.
JA, Ortu~ no A et al. Phenolic diterpenes, flavones, and rosmarinic 24 Serra AT, Rocha J, Sepodes B, Matias AA, Feliciano RP, de Carv-
acid distribution during the development of leaves, flowers, stems, alho A et al. Evaluation of cardiovascular protective effect of dif-
and roots of Rosmarinus officinalis. Antioxidant activity. J Agric ferent apple varieties - Correlation of response with composition.
Food Chem 2003;51:4247–53. Food Chem 2012;135:2378–86.
4 Nakazawa T, Ohsawa K. Metabolism of rosmarinic acid in rats. J 25 Ou B, Hampsch-Woodill M, Flanagan J, Deemer EK, Prior RL,
Nat Prod 1998;61:993–6. Huang D. Novel fluorometric assay for hydroxyl radical prevention
5 Baba S, Osakabe N, Natsume M, Terao J. Orally administered ros- capacity using fluorescein as the probe. J Agric Food Chem
marinic acid is present as the conjugated and/or methylated forms 2002;50:2772–7.
in plasma, and is degraded and metabolized to conjugated forms of 26 Freitas M, Porto G, Lima JL, Fernandes E. Isolation and activation
caffeic acid, ferulic acid and m-coumaric acid. Life Sci of human neutrophils in vitro: the importance of the anticoagulant
2004;75:165–78. used during blood collection. Clin Biochem 2008;41:570–5.
6 Konishi Y, Kobayashi S. Transepithelial transport of rosmarinic 27 Freitas M, Porto G, Lima JL, Fernandes E. Optimization of experi-
acid in intestinal Caco-2 cell monolayers. Biosci Biotechnol Bio- mental settings for the analysis of human neutrophils oxidative
chem 2005;69:583–91. burst in vitro. Talanta 2009;78:1476–83.
7 Debersac P, Vernevaut MF, Amiot MJ, Suschetet M, Siess MH. 28 Freitas M, Lima JL, Fernandes E. Optical probes for detection and
Effects of a water-soluble extract of rosemary and its purified com- quantification of neutrophils’ oxidative burst: a review. Anal Chim
ponent rosmarinic acid on xenobiotic-metabolizing enzymes in rat Acta 2009;649:8–23.
liver. Food Chem Toxicol 2001;39:109–17. 29 Fernandes E, Costa D, Marques AP, Reis RL, Lima JLFC. Inhibi-
8 Conner EM, Grisham MB. Inflammation, free radicals, and antioxi- tion of human neutrophil oxidative burst by pyrazolone derivatives.
dants. Nutrition 1996;4:274–7. Free Radic Biol Med 2006;40:632–40.

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
412 JOAO ROCHA ET AL.

30 Vogel HG. Drug Discovery and Evaluation: Pharmacological 49 Elias-Miro M, Jimenez-Castro MB, Rodes J, Peralta C. Current
Assays. Springer-Verlag and Heidelberg GmbH & Co., Berlin, knowledge on oxidative stress in hepatic ischemia/reperfusion. Free
Germany, 2002;751–72. Radic Res 2013;8:555–68.
31 Bignotto L, Rocha J, Sepodes B, Eduardo-Figueira M, Pinto R, 50 Osakabe N, Yasuda A, Natsume M, Sanbongi C, Kato Y, Osawa
Chaud M et al. Anti-inflammatory effect of lycopene on carra- T et al. Rosmarinic acid, a major polyphenolic component of Peri-
geenan-induced paw oedema and hepatic ischaemia-reperfusion in lla frutescens, reduces lipopolysaccharide (LPS)-induced liver
the rat. Br J Nutr 2009;102:126–33. injury in D-galactosamine (D-GalN)-sensitized mice. Free Radic
32 Sepodes B, Maio R, Pinto R, Sharples E, Oliveira P, McDonald M Biol Med 2002;33:798–806.
et al. Recombinant human erythropoietin protects the liver from 51 Dahiya P. Burns as a model of SIRS. Front Biosci (Landmark Ed)
hepatic ischemia-reperfusion injury in the rat. Transpl Int 2009;1:4962–7.
2006;19:919–26. 52 Dancey DR, Hayes J, Gomez M, Schouten D, Fish J, Peters W
33 Fischer AE, Jacobson KA, Rose J, Zeller R. Hematoxylin and et al. ARDS in patients with thermal injury. Intensive Care Med
eosin staining of tissue and cell sections. CSH Protoc 2008;2008: 1999;25:1231–6.
pdb.prot4986. 53 Herridge MS, Cheung AM, Tansey CM, Matte-Martyn A, Diaz-
34 Schreiber E, Matthias P, M€uller MM, Schaffner W. Rapid detec- Granados N, Al-Saidi F et al. One-year outcomes in survivors of
tion of octamer binding proteins with ‘mini-extracts’, prepared the acute respiratory distress syndrome. N Engl J Med
from a small number of cells. Nucleic Acids Res 1989;17:6419. 2003;348:683–93.
35 Fernandes A, Falc~ao AS, Silva RF, Gordo AC, Gama MJ, Brito MA 54 Rodriguez JL, Miller CG, Garner WL, Till GO, Guerrero P, Moore
et al. Inflammatory signalling pathways involved in astroglial activa- NP et al. Correlation of the local and systemic cytokine response
tion by unconjugated bilirubin. J Neurochem 2008;96:1667–79. with clinical outcome following thermal injury. J Trauma
36 Barateiro A, Vaz AR, Silva SL, Fernandes A, Brites D. ER stress, 1993;34:684–95.
mitochondrial dysfunction and calpain/JNK activation are involved 55 Ipaktchi K, Mattar A, Niederbichler AD, Hoesel LM, Vollmann-
in oligodendrocyte precursor cell death by unconjugated bilirubin. shauser S, Hemmila MR et al. Attenuating burn wound inflamma-
Neuromolecular Med 2012;14:285–302. tory signaling reduces systemic inflammation and acute lung
37 Silva SL, Vaz AR, Barateiro A, Falc~ao AS, Fernandes A, Brito injury. J Immunol 2006;177:8065–71.
MA et al. Features of bilirubin-induced reactive microglia: from 56 Abdullahi A, Amini-Nik S, Jeschke MG. Animal models in burn
phagocytosis to inflammation. Neurobiol Dis 2010;40:663–75. research. Cell Mol Life Sci 2014;71(17):3241–55 (in press).
38 Di Rosa M, Giroud JP, Willoughby DA. Studies on the mediators 57 Brachkova MI, Marques P, Rocha J, Sepodes B, Duarte MA, Pinto
of the acute inflammatory response induced in rats in different sites JF. Alginate films containing Lactobacillus plantarum as wound
by carrageenan and turpentine. J Pathol 1971;104:15–29. dressing for prevention of burn infection. J Hosp Infect
39 Khattab MM. Tempol, a membrane-permeable radical scavenger, 2011;79:375–7.
attenuates peroxynitrite- and superoxide anion-enhanced carra- 58 Jeschke MG, Einspanier R, Klein D, Jauch KW. Insulin attenuates
geenan-induced paw edema and hyperalgesia: a key role for super- the systemic inflammatory response to thermal trauma. Mol Med
oxide anion. Eur J Pharmacol 2006;548:167–73. 2002;8:443–50.
40 Cuzzocrea S, Zingarelli B, Hake P, Salzman AL, Szabo C. Antiin- 59 Matute-Bello G, Downey G, Moore BB, Groshong SD, Matthay
flammatory effects of mercaptoethylguanidine, a combined inhibi- MA, Slutsky AS et al. An official American Thoracic Society
tor of nitric oxide synthase and peroxynitrite scavenger, in workshop report: features and measurements of experimental
carrageenan-induced models of inflammation. Free Radic Biol Med acute lung injury in animals. Am J Respir Cell Mol Biol
1998;24:450–9. 2011;44:725–38.
41 Landino LM, Crews BC, Timmons MD, Morrow JD, Marnett LJ. 60 Datta G, Fuller BJ, Davidson BR. Molecular mechanisms of liver
Peroxynitrite, the coupling product of nitric oxide and superoxide, ischemia reperfusion injury: insights from transgenic knockout
activates prostaglandin biosynthesis. Proc Natl Acad Sci USA models. World J Gastroenterol 2013;11:1683–98.
1996;93:15069–74. 61 Grommes J, Soehnlein O. Contribution of neutrophils to acute lung
42 D’Acquisto F, May MJ, Ghosh S. Inhibition of nuclear factor injury. Mol Med 2011;17:293–307.
kappa B (NF-B): an emerging theme in anti-Inflammatory thera- 62 Lee WL, Downey GP. Neutrophil activation and acute lung injury.
pies. Mol Interv 2002;2:22–35. Curr Opin Crit Care 2001;7:1–7.
43 Thiemermann C, McDonald MC, Cuzzocrea S. The stable nitrox- 63 Edens HA, Parkos CA. Neutrophil transendothelial migration and
ide, tempol, attenuates the effects of peroxynitrite and oxygen- alteration in vascular permeability: focus on neutrophil-derived
derived free radicals. Crit Care Med 2001;29:223–4. azurocidin. Curr Opin Hematol 2003;10:25–30.
44 Cuzzocrea S, Thiemermann C, Salvemini D. Potential therapeutic 64 Fry DE. Sepsis, systemic inflammatory response, and
effect of antioxidant therapy in shock and inflammation. Curr Med multiple organ dysfunction: the mystery continues. Am Surg
Chem 2004;11:1147–62. 2012;78:1–8.
45 Naito Y, Takano H, Yoshikawa T. Oxidative stress-related mole- 65 Parks WC, Wilson CL, Ló pez-Boado YS. Matrix metalloproteinas-
cules as a therapeutic target for inflammatory and allergic diseases. es as modulators of inflammation and innate immunity. Nat Rev
Curr Drug Targets Inflamm Allergy 2005;4:511–5. Immunol 2004;4:617–29.
46 Kaplan M, Mutlu EA, Benson M, Fields JZ, Banan A, Keshavarz- 66 Davey A, McAuley DF, O’Kane CM. Matrix metalloproteinases in
ian A. Use of herbal preparations in the treatment of oxidant-medi- acute lung injury: mediators of injury and drivers of repair. Eur
ated inflammatory disorders. Complement Ther Med 2007;15: Respir J 2011;38:959–70.
207–16. 67 Qiu Z, Hu J, Van den Steen PE, Opdenakker G. Targeting matrix
47 Moreno S, Scheyer T, Romano CS, Vojnov AA. Antioxidant and metalloproteinases in acute inflammatory shock syndromes. Comb
antimicrobial activities of rosemary extracts linked to their poly- Chem High Throughput Screen 2012;15:555–70.
phenol composition. Free Radic Res 2006;40:223–31. 68 Vandenbroucke RE, Dejonckheere E, Libert C. A therapeutic role
48 Erkan N, Ayranci G, Ayranci E. Antioxidant activities of rosemary for matrix metalloproteinase inhibitors in lung diseases? Eur Respir
(Rosmarinus officinalis L.) extract, blackseed (Nigella sativa L.) J 2011;38:1200–14.
essential oil, carnosic acid, rosmarinic acid and sesamol. Food 69 Baud V, Karin M. Is NF-kappaB a good target for cancer therapy?
Chem 2008;110:76–82. Hopes and pitfalls. Nat Rev Drug Discov 2012;8:33–40.

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)
EFFECT OF ROSMARINIC ACID IN INFLAMMATION 413

70 Ghosh S, Hayden MS. New regulators of NF-kappaB in inflamma- role of reactive oxygen species and inflammation. Biomed Res Int
tion. Nat Rev Immunol 2008;8:837–48. 2013;2013:845037.
71 Oeckinghaus A, Hayden MS, Ghosh S. Crosstalk in NF-jB signal- 74 Rosanna DP, Salvatore C. Reactive oxygen species,
ing pathways. Nat Immunol 2011;12:695–708. inflammation, and lung diseases. Curr Pharm Des 2012;18:3889–
72 Minghetti L. Role of inflammation in neurodegenerative diseases. 900.
Curr Opin Neurol 2005;18:315–21. 75 Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB. Oxidative
73 Lin CP, Lin FY, Huang PH, Chen YL, Chen WC, Chen HY et al. stress, inflammation, and cancer: how are they linked? Free Radic
Endothelial progenitor cell dysfunction in cardiovascular diseases: Biol Med 2010;49:1603–16.

© 2014 Nordic Association for the Publication of BCPT (former Nordic Pharmacological Society)

You might also like