Acessibilidade / Reportar erro

In vivo anti-inflammatory and antinociceptive effects, and in vitro antioxidant, antiglycant and anti-neuroinflammatory actions of Syzygium malaccense

Abstract

Syzygium malaccense is popularly used to treat inflammation and pain-related ailments. The species was assessed regarding its antioxidant, antiglycant, anti-inflammatory, including anti-neuroinflammatory, and antinociceptive activities. Different models were employed to measure S. malaccense extract (ESM) antioxidant activity. The antiglycant activity was determined using the glucose-induced protein glycation model. LPS-induced neuroinflammation on murine BV-2 microglial cell line was used for anti-neuroinflammatory activity evaluation. The croton oil-induced ear edema test was accomplished to evaluate the in vivo anti-inflammatory activity. Acetic acid-induced writhing together with formalin-induced paw licking assays were performed to evaluate the antinociceptive potential. Finally, the chemical characterization was accomplished by a UHPLC-MS analysis. ESM presented relevant antioxidant and antiglycant activity. NO production by BV-2 cells was reduced, indicating the relevant neuroprotective activity. ESM significantly decreased the mice ear edema induced by croton oil and the nociceptive stimulus induced by acetic acid and formalin by central and peripheral mechanisms. The flavonoids myricitrin, myricetin and quercetin were identified and, as far as we know, the alkaloid reserpine was reported in the species for the first time. The antioxidant and antiglycant potential of ESM, may be related to the in vivo anti-inflammatory and antinociceptive effects, and to the in vitro neuroinflammation inhibition.

Key words
Antiglycant; antioxidant; inflammation; oxidative stress; pain

INTRODUCTION

The inflammatory process is a severe response triggered by tissues to restore homeostasis after a harmful stimulus. Inflammation often causes noticeable symptoms, such as pain, which is one of the main reasons for medical sessions (Treed et al. 2019TREED RD ET AL. 2019. Chronic pain as a symptom or a disease. Pain 160: 19-27. https://doi.org/ 10.1097/j.pain.0000000000001384.). The Global Burden of Diseases recognizes chronic pain as one of the leading causes of disability worldwide, which encourages national governments to consider pain management a public health priority and generate national strategies to challenge this issue (Fayaz et al. 2016FAYAZ A, CROFT P, LANGFORD RM, DONALDSON LJ & JONES GT. 2016. Prevalence of chronic pain in the UK: a systematic review and meta-analysis of population studies. BMJ Open 6: e010364. https://doi.org/10.1136/bmjopen-2015-010364.). Pain reduces life quality and represents a significant risk for developing mental health disorders and suicidality, characterized by ideation, plans, and attempts to suicide (Campbell et al. 2015CAMPBELL G, DARKE S, BRUNO R & DEGENHARDT L. 2015. The prevalence and correlates of chronic pain and suicidality in a nationally representative sample. Aust N Z J Psychiatry 49: 803-811. https://doi.org/10.1177/0004867415569795.).

Furthermore, neurodegenerative disorders, including Alzheimer, Parkinson, Huntington, and multiple sclerosis, have emerged along with the increase of life expectancy worldwide (Zhang et al. 2010ZHANG C, BROWNE A, CHILD D, DIVITO JR, STEVENSON JA & TANZI RE. 2010. Loss of function of ATXN1 increases amyloid beta-protein levels by potentiating beta-secretase processing of beta-amyloid precursor protein. J Biol Chem 285: 8515-8526. https://doi.org/10.1074/jbc.M109.079079., Elmann et al. 2011ELMANN A, MORDECHAY S, ERLANK H, TELERMAN A, RINDNER M & OFIR R. 2011. Anti-neuroinflammatory effects of the extract of Achillea fragrantissima A. BMC Complement Altern Med 11: 1198. https://doi.org/10.1186/1472-6882-11-98.). Those diseases are related to a neuroinflammatory process and the excessive production of pro-inflammatory mediators by brain microglial cells (Elmann et al. 2011ELMANN A, MORDECHAY S, ERLANK H, TELERMAN A, RINDNER M & OFIR R. 2011. Anti-neuroinflammatory effects of the extract of Achillea fragrantissima A. BMC Complement Altern Med 11: 1198. https://doi.org/10.1186/1472-6882-11-98., Kim et al. 2018KIM S, BAEK SK & SONG KB. 2018. Physical and antioxidant properties of alginate films prepared from Sargassum fulvellum with black chokeberry extract. Food Packag Shelf Life 18: 157-163. https://doi.org/10.1186/1472-6882-11-98.).

Oxidative stress, several chemical mediators and cellular components are involved in the inflammatory response, playing essential roles in the physiopathology of various high prevalence diseases, including rheumatoid arthritis, atherosclerosis, and asthma (Bhagavan et al. 2013BHAGAVAN NB, ARUNACHALAM S, DHASARATHAN P & KANNAN ND. 2013. Evaluation of anti-inflammatory activity of Indigofera aspalathoides Vahl in Swiss albino mice. J Pharm Res 6: 350-354. https://doi.org/10.1016/j.jopr.2013.02.018.). There is also evidence that oxidative stress is associated with neurotoxic mechanisms involved in the pathogenesis of neurodegenerative disorders (Neal & Richardson 2018NEAL M & RICHARDSON JR. 2018. Time to get personal: A framework for personalized targeting of oxidative stress in neurotoxicity and neurodegenerative disease. Curr Opin Toxicol 7: 127-132. https://doi.org/10.1016/j.cotox.2018.02.003.). The brain oxidative stress may induce the formation of advanced glycation end products (AGEs), resulting from a non-enzymatic reaction that binds sugars to peptides or proteins. AGEs formation enhances the oxidative stress, inducing a positive feedback loop and favors the deposition of proteins in neurons, including amyloid-β, tau, α-synuclein, and prions, which are related to the neuronal cell death and, consequently, to the development of neurodegenerative diseases (Li et al. 2012LI J, LIU D, SUN L, LU Y & ZHANG Z. 2012. Advanced glycation end products and neurodegenerative diseases: mechanisms and perspective. J Neurol Sci 317: 1-5. https://doi.org/10.1016/j.jns.2012.02.018.).

In this context, several plants are used in traditional medicine for inflammation and pain-related ailments., including Syzygium malaccense (L.) Merr. & L. M. Perry (Myrtaceae), basionym Eugenia malaccensis L. This plant is native from Malaysia, Indonesia, Vietnam, and Thailand. However, it is commonly found in other tropical regions, as in the northern, northeastern, and southeastern regions of Brazil, where it is popularly known as “jambo” and “jambo vermelho” (Nunes et al. 2016NUNES PC, AQUINO JS, ROCKENBACH II & STAMFORD TLM. 2016. Physico-chemical characterization, bioactive compounds and antioxidant activity of malay apple [Syzygium malaccense (L.) Merr. & L.M. Perry]. PLoS ONE 11: 1-11. https://doi.org/10.1371/journal.pone.0158134., Fernandes & Rodrigues 2018FERNANDES FAN & RODRIGUES S. 2018. Jambo - Syzygium malaccense. Exotic Fruits 245-249. https://doi.org/10.1016/B978-0-12-803138-4.00031-9.). Different parts of this plant have been used in traditional medicine to treat various diseases, including dysentery, fever, bronchitis, skin diseases, and abdominal pain. It is also used as a diuretic, anti-inflammatory, antiviral and antifungal (Adebayo et al. 2015ADEBAYO AH, OGUNDARE OC & ADEGBITE OS. 2015. Sub-acute evaluation of extract of Syzygium malaccense in albino rats. J Med Plant Res 9: 60-71. https://doi.org/10.3923/rjmp.2015.60.71., Dustan 1997, Pedrollo et al. 2016PEDROLLO CT, KINUPP VF, SHEPARD-JR G & HEINRICH M. 2016. Medicinal plants at Rio Jauaperi, Brazilian Amazon: Ethnobotanical survey and environmental conservation. J Ethnopharmacol 186: 111-124. https://doi.org/10.1016/j.jep.2016.03.055., Whistler & Elevitch 2006WHISTLER WA & ELEVITCH CR. 2006. Syzygium malaccense. In: Elevitch CR (Ed), Species Profiles for Pacific Island Agroforestry. Holualoa, Hawai: Permanent Agriculture Resources (PAR), p. 1-13.).

The antioxidant and hypoglycemic potential were reported for the alcoholic extract obtained from the leaves of S. malaccense (Arumugam et al. 2016ARUMUGAM B, MANAHARAN T, HENG CK, KUPPUSAMY UR & PALANISAMY UD. 2016. Antioxidant and antiglycemic potentials of a standardized extract of Syzygium malaccense. LWT - Food Sci Technol 59: 707-712. https://doi.org/10.1016/j.lwt.2014.06.041., Ramadhania et al. 2017RAMADHANIA ZM, INSANU M, GUNARTI NS, WIRASUTISNA KR, SUKRASNO S & HARTATI R. 2017. Antioxidant activity from ten species of Myrtaceae. Asian J Pharm Clin Res Special Iissue May. https://doi.org/10.22159/ajpcr.2017.v10s2.19470.). The ethanolic extract presented a significant topical inhibitory effect against the ethyl phenyl propiolate-induced rat ear edema challenge and a promising in vitro inhibition of the enzyme cyclooxygenase-1 (Dunstan 1997DUNSTAN CA. 1997. Evaluation of some Samoan and Peruvian medicinal plants by prostaglandin biosynthesis and rat ear oedema assays. J Ethnopharmacol 57: 35-56. https://doi.org/10.1016/s0378-8741(97)00043-3.). Glycosylated flavonoids as myricetin derivatives (Arumugam et al. 2016ARUMUGAM B, MANAHARAN T, HENG CK, KUPPUSAMY UR & PALANISAMY UD. 2016. Antioxidant and antiglycemic potentials of a standardized extract of Syzygium malaccense. LWT - Food Sci Technol 59: 707-712. https://doi.org/10.1016/j.lwt.2014.06.041.), catechins, quercetin, and carotenoids were already identified in the leaves (Batista 2017BATISTA AG. 2017. Red-jambo (Syzygium malaccense): bioactive compounds in fruits and leaves. LWT - Food Sci Technol 76: 284-291. https://doi.org/10.1016/j.lwt.2016.05.013.).

The present study investigated the potential of the methanolic extract of S. malaccense leaves (ESM) in treating painful and inflammatory conditions, including neuroinflammation. In addition, its antioxidant and antiglycant activities were assessed, as oxidative stress and advanced glycation end products may be involved in the pathophysiology of brain inflammation. Besides, ESM chemical characterization was performed.

MATERIALS AND METHODS

Plant material

The leaves of S. malaccense were collected in Rio Novo, Minas Gerais State, Brazil (21°26’03.6”S 43°06’41.8”W). A voucher specimen (CESJ 46600) was deposited at Leopoldo Krieger Herbarium of Federal University of Juiz de Fora. The plant name has been checked with http://www.the plant list.org, accessed in July 2020.

The plant material was dried at 40 °C, and the extraction process was performed by static maceration at room temperature with methanol until exhaustion. Finally, the extract was concentrated under reduced pressure using a rotary evaporator to obtain the methanolic extract (ESM), and stored in a refrigerator at 4 °C.

Chemicals

Indomethacin was purchased from Las Casas (Juiz de Fora, MG, Brazil). Morphine was obtained from Cristália (Itapira, SP, Brazil). Animal commercial chow was from Nuvital® (Colombo, PR, Brazil). Croton oil, Tween 80, quercetin, 1,1-diphenyl-2-picrylhydrazyl (DPPH), β-carotene, linoleic acid, bovine serum albumin (BSA), fetal bovine serum (FBS), E. coli serotype 111:B4 lipopolysaccharide (LPS), 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), aminoguanidine and fructose were from Sigma-Aldrich (St. Louis, MO, USA). Sodium phosphate was from Labsynth (Diadema, SP, Brazil), and ammonium molybdate was from Vetec (Rio de Janeiro, RJ, Brazil). Dulbecco´s modified Eagle´s medium (DMEM) and antibiotic-antimycotic solution were acquired from Thermo-Fischer Scientific (Waltham, MA, USA). All other reagents were of the highest quality available.

In vitro antioxidant activity

β-carotene/linoleic acid bleaching assay

Antioxidant activity was determined using the β-carotene bleaching assay (Ismail et al. 2004ISMAIL A, MARJAN ZM & FOONG CW. 2004. Total antioxidant activity and phenolic content in selected vegetables. Food Chem 87: 581-586. https://doi.org/10.1016/j.foodchem.2004.01.010.) with slight modifications. A stock solution of the β-carotene-linoleic acid mixture was prepared as follows: 50 µL β-carotene (10 mg/mL in chloroform), 20 µL linoleic acid, and 265 µL Tween 40 were dissolved in 1 mL of chloroform. The solvent was then entirely evaporated using nitrogen gas, and 40 mL of oxygen saturated water was added with vigorous shaking. After, 250 µL of this mixture were dispersed to a 96-well plate. Finally, 10 μL of blank, ESM, and quercetin (final concentration 1.20-38.5 μg/mL), used as the reference standard, were added. The emulsion system was incubated for 2 h at 45 °C, and the absorbance was read at 470 nm. The test was performed in triplicate. IC50 values were calculated and indicated the concentration of extract required to inhibit 50% of oxidation.

DPPH radical scavenging test

The radical 1,1-diphenyl-2-picryl-hydrazyl (DPPH) was used to determine ESM free radical-scavenging activity (Blois 1958BLOIS MS. 1958. Antioxidant determination by the use of stable free radical. Nature 181: 1199-2000. https://doi.org/10.1038/1811199a0.). One hundred microliters of ESM diluted in methanol were added to a 96-well microplate. Successive (1:2) dilution with methanol was then performed (250 - 0.98 μg/mL). Methanol solution of DPPH 20 μg/mL was added to each well. The microplate was incubated in the dark for 30 min, and the absorbance was measured at 517 nm. The experiment was carried out in triplicate. Quercetin and ascorbic acid were used as reference compounds. IC50 values were calculated and indicated the concentration of extract required to scavenge 50% of DPPH free radicals.

Phosphomolybdenum assay

This test was accomplished as described by Prieto et al. (1999)PRIETO P, PINEDA M & AGUILAR M. 1999. Spectrophotometric quantitation of antioxidant capacity through the formation of a phosphomolybdenum complex: specific application to the determination of vitamin E. Anal Biochem 269: 337-341. https://doi.org/10.1006/abio.1999.4019.. The phosphomolybdenum is produced after the reaction between Na3PO4 (28 mL, 0.1mol/L) with (NH4)6Mo7O24.4 H2O (12 mL, 0.03 mol/L) and H2SO4 (20 mL, 3 mol/L) in water (sufficient quantity to 100 mL). Two milliliters of the reactive solution was added to 200 μL of ESM or rutin (200 μg/mL), used as the reference drug. The mixture was incubated in a water bath at 95 °C for 90 min. The absorbance was measured at 695 nm. The test was performed in triplicate. The total antioxidant capacity (TAC) was expressed as milligrams of ESM equivalent to 1 mg of ascorbic acid, using a seven-point standard calibration curve.

Ferric reducing antioxidant power assay (FRAP)

The ESM reducing power was determined by FRAP assay described by Oyaizu (1986)OYAIZU M. 1986. Studies on product of browning reaction prepared from glucose amine. Jap J Nut Diet 44: 307-315. https://doi.org/10.5264/eiyogakuzashi.44.307. at different concentrations (serial dilution of 3.35-53.64 µg/mL). Firstly, ESM was diluted in phosphate buffer pH 6.6 and potassium ferrocyanide [K3Fe(CN)6] 1% and incubated at 50 °C for 20 min. After that, trichloroacetic acid 10% was added, and the mixture was centrifuged at 3000 rpm for 10 min. The supernatant was diluted in 2.5 mL water and 0.5 mL FeCl3 0.1%, and the absorbance was measured using a spectrophotometer (700 nm). Ascorbic acid was used as the reference compound. This test was performed in triplicate, and the EC50 was calculated using the mean of five different concentrations.

Thiobarbituric acid reactive substances assay (TBARS)

The TBARS assay was used to verify ESM ability to inhibit lipid peroxidation in ground beef, as described by Uchiyama & Mihara (1978)UCHIYAMA M & MIHARA M. 1978. Determination of malonaldehyde precursor in tissues by thiobarbituric acid test. Anal Biochem 86: 271-278. https://doi.org/10.1016/0003-2697(78)90342-1., with some modifications. Briefly, 100 g of ground beef and 67 mL of distilled and deionized water were mixed with 7.5, 15, or 30 mg of ESM dissolved in methanol. A solution containing beef, water, and methanol was used as the control, and butylated hydroxytoluene (BHT) 7.5, 15, or 30 mg diluted in methanol was used as the reference compound. These mixtures were blended until a smooth homogenate was formed and transferred to amber jars, stored at 5 °C for 5 days. The beef oxidation was then indirectly measured by a calibration curve prepared using the malonaldehyde (MDA) standard, reacting with the TBA/phosphoric acid solution. The measurement was accomplished using the absorbance spectrum of 535 nm. The results were expressed as µM of MDA formed by the reaction.

In vitro antiglycant activity

The in vitro antiglycant activity was determined using glucose-induced protein glycation models. The method was performed as previously described (Farsi et al. 2008FARSI DA, HARRIS CS, REID L, BENNET SAL, HADDAD PS, MARTINEAU LC & ARNASON JT. 2008. Inhibition of non-enzymatic glycation by silk extracts from a Mexican land race and modern inbred lines of maize (Zea mays). Phytother Res 22: 108-112. https://doi.org/10.1002/ptr.2275.) with some modifications. The ESM and aminoguanidine were evaluated at concentrations of 375-12.5 μg/mL in a medium containing BSA (2.5 mg/mL), glucose (0.2 M), sodium azide (0.6 g/L) and sodium phosphate buffer (pH 7.4). After the incubation time at 37 °C, the amount of fluorescent AGEs formed was determined using a fluorimeter after one, two, and ten days. The fluorescent intensity was measured at 330 nm (excitation) and 410 nm (emission). The antiglycant activity was expressed as the mean ± S.E.M. of AGE formation percentage compared to the vehicle.

In vitro anti-neuroinflammatory activity on murine BV-2 microglial cell line

Cell culture

Murine BV-2 microglial cell line was used to evaluate the anti-neuroinflammatory activity of ESM. The cells were maintained in DMEM enriched with 10% FBS and 1% of antibiotic solution (10.000 units/mL of penicillin, 10.000 µg/mL of streptomycin, and 25 µg/mL of Gibco Amphotericin B) at 37 °C, 95% humidity and 5% CO2. The cultures were subcultivated in 25 and 75 cm2 bottles, with 0.05% trypsin diluted in ethylenediamine tetraacetic acid (EDTA).

Cytotoxic activity on BV-2 cells

To evaluate whether ESM is toxic to BV-2 cells, the MTT assay (Mosmann 1983MOSMANN T. 1983. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods 65: 55-63. https://doi.org/10.1016/0022-1759(83)90303-4.) was accomplished. Therefore, 4 x 104 cells/well were seeded in 96-well microplate and incubated overnight. Then, ESM 50 μg/mL was added, and the plate was incubated for 3 and 24 h at 37 °C, 100% humidity, and 5% CO2. After the incubation time, the wells were aspirated, and 200 μL of MTT (0.5 mg/mL) diluted in DMEM were added to each well. Finally, after 2 h of the incubation period, the formazan crystals were dissolved in dimethyl sulfoxide (DMSO)/ethanol (1:1). The control groups consisted of 0.3% DMSO or DMEM medium. The optical density was read at 570 nm. The test was performed in quintuplicate, and the results were expressed as mean ± S.E.M. of three independent experiments.

In vitro LPS-induced neuroinflammation

This test was accomplished according to Wang-Yang et al. (2014)WANG-YANG W, WU Y, HUANG H, HE C, LI W, WANG H, CHEN H & YIN Y. 2014. Biochanin A attenuates LPS-induced pro-inflammatory responses and inhibits the activation of the MAPK pathway in BV2 microglial cells. Int. J Mol Med 35: 391-398. https://doi.org/10.3892/ijmm.2014.2020.. Therefore, 4 x 104 cells/well were seeded in 96-well microplate and incubated overnight. Then, the wells were aspirated, and 50 μg/mL of ESM, diluted in DMEM enriched with 2% FBS, were added. After 2 h, bacterial lipopolysaccharide (LPS) 1 μg/mL was also added, and the mixture was incubated for 24 h. The induction control consisted of DMEM with 2% FBS and LPS. The negative control, DMEM with 2% FBS, was used to evaluate the cell growth. After that, the supernatants were transferred to a sterile 96-well microplate for nitric oxide (NO) dosage. Then, 200 μL of MTT reagent (0.5 mg/mL) diluted in DMEM were added to each well, and the mixtures were incubated for 2 h. The insoluble formazan crystals were dissolved using 200 μL of DMSO/ethanol (1:1) solution per well. The optical density was evaluated at 570 nm. The NO level in cell cultures was measured using the Griess reaction (Schmidt & Kelm 1996SCHMIDT HW & KELM M. 1996. Determination of nitrite and nitrate by the Griess reaction. In: FEELISCH M & STAMLER JS (Eds.), Methods in Nitric Oxide Research, New York: J Wiley & Sons Ltd., New York, USA, p. 491-497.). The test was performed in quintuplicate, and the results were expressed in the inhibition percentage compared to the induction control. The mean ± S.E.M. of three independent experiments was used.

In vivo anti-inflammatory and antinociceptive activities

Animals

Male Swiss mice (20-30 g, approx. 30 days old) were bred at the Center of Reproductive Biology of Federal University of Juiz de Fora. The animals were maintained in a room with 12 h/12 h dark/light cycle under standard temperature (22 °C) with water and food ad libitum; however, mice were fasted for 12 h before each experiment. Each group contained 8 animals. All experimental procedures followed the Ethical Principles of Animal Research adopted by the Brazilian College of Animal Experimentation (COBEA – CEUA/UFJF Protocols n°009/2009, 013/2013, 016/2013, 021/2013 and 028/2014)

Croton oil-induced ear edema test

The anti-inflammatory activity of ESM was evaluated as described by Schiantarelli and collabs (Schiantarelli et al. 1982SCHIANTARELLI P, CADEL S, ACERBI D & PAVESI L. 1982. Anti-inflammatory activity and bioavailability of percutaneous piroxican. Drug Res 32: 230-235.). Firstly, animals were distributed in five groups and orally treated by gavage with ESM 50, 100, or 300 mg/kg, indomethacin 10 mg/kg (used as reference drug), or vehicle (12% Tween 80 diluted in NaCl 0.9%, v/v). All solutions were administered at 10 mL/kg. After 60 min, 20 μL of a fresh solution of 2.5% croton oil dissolved in acetone (v/v) was topically applied in the inner surface of the right ear, and the same volume of acetone (vehicle) was administered on the inner surface of the left ear of each mouse. Six hours after the beginning of the experiment, the animals were euthanized, and 6 mm diameter ear punch biopsies were obtained and individually weighed in an analytical balance. The weight difference between the right (inflamed) and the left (non-inflamed) ear biopsies was used to measure edema and, consequently, the inflammatory process.

Acetic acid-induced writhing test

The evaluation of the antinociceptive action of ESM was performed using the well-established acetic acid-induced writhing test (Koster et al. 1959KOSTER R, ANDERSON M & DE BEER EJ. 1959. Acetic acid for analgesic screening. Fed Proc 18: 412-418.). Five groups of animals were used. Mice were orally treated with ESM 50, 100, or 300 mg/kg, indomethacin 10 mg/kg, used as the reference drug, or vehicle (12% Tween 80 diluted in NaCl 0.9%, v/v). All treatments were administered at 10 mL/kg. After 60 min, mice received i.p. injection of acetic acid 0.6%, 10 mL/kg. The number of writhings (constriction of the abdominal wall combined with the extension of the hind paws) was counted for 30 min and indicated the level of mice nociceptive reaction.

Formalin-induced paw licking test

The formalin test was used to verify whether the ESM antinociceptive effect is related to a central or peripheral anti-inflammatory activity (Hunskaar et al. 1985HUNSKAAR S, FASMER OB & HOLE K. 1985. The formalin test in mice, a useful technique for evaluating mild analgesia. J Neurosci Methods 14: 69-76. https://doi.org/10.1016/0165-0270(85)90116-5.). Firstly, animals were separated in six groups. Mice were orally pretreated with 10 mL/kg of ESM 50, 100 or 300 mg/kg, indomethacin 10 mg/kg, which was used as a reference drug that acts peripherally, or vehicle (12% Tween 80 diluted in NaCl 0.9%, v/v). Besides, morphine 7.5 mg/kg (10 mL/kg) was i.p. injected to be used as a reference antinociceptive drug with central mechanisms. After 60 min, 20 mL of 2% formalin prepared in saline was injected into each mouse’s right hind paw’s sup-plantar tissue. The licking paw time was timed during 5 min (first phase of the nociceptive stimulus) and between 15 and 30 min (second phase of the nociceptive stimulus) after the formalin injection.

Chemical characterization

UHPLC-MS analysis

The instrument consisted of an ultra-high pressure liquid chromatography (UHPLC) Shimadzu Nexera attached to a high-resolution mass spectrometer, a quadrupole time of flight (QTOF) Bruker Maxis, with an electrospray ionization operated in a positive mode. The chromatographic separation was achieved in a C18 Shim-pack XR-ODSIII (150 x 2.0 mm, 2.2 μm) at a controlled temperature of 40 oC, and the injection volume was set to 5 μL.

The mobile phase consisted of 0.1% formic acid in water as reservoir A and 0.1% formic acid in acetonitrile as reservoir B at a constant flow rate of 200 μL/min. The mobile phase was delivered according to the following elution program: 5.0% B (0.0 – 5.0 min); 5.0-100.0% B (5.0 – 45.0 min); 100% B (45.0 – 50.0); and one more minute to restore the column to the initial mobile phase condition. Samples were introduced into the interface through a heated nebulizer probe set at 200 °C. The optimized parameters for electrospray ionization (ESI) were a gas flow of 8 L/min, a nebulizer of 2.0 bar, a capillary voltage of 4.5 kV, and a gas temperature of 200 oC, with nitrogen used as the drying gas. Compass software version 1.5 was used to obtain data acquisition and quantification.

HPLC-UV analysis

This technique was used to confirm the presence of the alkaloid reserpine in ESM. Reserpine standard (0.15 mg/mL), ESM (2 mg/mL), and the mixture of both samples (Reserpine:ESM – 1:100) were injected at room temperature (25 °C) in the high-pressure liquid chromatography (HPLC) instrument attached to UV detector Agilent 1200 Series (Agilent, Santa Clara, CA). The mobile phase was a gradient consisted of water and acetonitrile 5.0% - 70% (0.0 – 10.0 min), then acetonitrile 70% - 80% (10.01 – 30.0 min). The volume injection was set to 20 µL. Agilent XDB-C18 column, the flow rate at 0.8 mL/min, and detection at 230 nm were used.

Statistical analysis

All antioxidant results were expressed as mean ± S.D. (standard deviation), except for TBARS assay. The software GraFit Data Analysis® 5.0 was used to calculate the IC50 in the DPPH scavenging assay and β-carotene/linoleic acid bleaching method. Microsoft Excel® 2007 was used to analyze the FRAP assay. The comparison between groups was assessed by two-way ANOVA, followed by the Tukey test in TBARS assay (mean ± S.E.M - standard error of mean). Values were expressed as mean ± S.E.M. One-way ANOVA followed by the Tukey test was used for all other experiments. The software GraphPad Prism® 7.0 was used for the statistical analysis. p values < 0.05 were considered significant.

RESULTS

Antioxidant activity

The Table I summarizes the IC50 values and total antioxidant capacity obtained for ESM in DPPH scavenging, β-carotene/linoleic acid bleaching and FRAP assays. ESM inhibited lipid peroxidation on days 4 and 5 at all tested concentrations in TBAR assays; the results were statistically comparable to BHT at 15 and 30 mg, in contrast to BHT 7.5 mg, which showed no activity (Figure 1).

Figure 1
ESM antioxidant activity in the TBARS assay. The ground beef oxidation was indirectly measured by a calibration curve prepared using the malonaldehyde (MDA) standard. The measurement was accomplished using the absorbance spectrum at 535 nm. The results were expressed as µM of MDA formed by the reaction. The comparison between groups was assessed by two-way ANOVA, followed by the Tukey test (mean ± S.E.M). Significant values: **p <0.01; ***p <0.001; ****p <0.0001 compared to the vehicle.
Table I
Antioxidant activity of Syzygium malaccense methanolic extract (ESM) evaluated by β-carotene/linoleic acid, DPPH, phosphomolybdenum, and FRAP assays.

Antiglycant activity

ESM showed impressively antiglycant activity, considerably reducing the in vitro formation of the BSA-glucose complex. ESM and aminoguanidine activities were similar. However, ESM showed more significant inhibition of AGE formation when statistically compared to the reference drug at some tested concentrations and time points throughout the experiment (Figure 2).

Figure 2
Effect of ESM at different concentrations (12.5-375μg/mL) on AGE formation. Fluorescent intensity was measured after (a) one, (b) two, and (c) ten days at 330 nm (excitation) and 410 nm (emission). The antiglycant activity was expressed as the mean ± S.E.M. of AGE formation percentage compared to the vehicle (100%). Significant values:****p <0.0001 compared to the vehicle; # means statistically equal to the reference drug (AMG- aminoguanidine) at the same concentration.

In vitro evaluation of the anti-neuroinflammatory activity of ESM in BV-2 microglial cell line

Firstly, it was verified if ESM interfered with BV-2 cell viability. ESM did not show cytotoxicity up to 100 μg/mL (data not shown). LPS was used to induce the inflammatory process on BV-2 cells at noncytotoxic levels (Figure 3), so that the cells induced and treated with ESM remained viable (as the control, C-). As shown in Figure 4, ESM significantly reduced NO production induced by LPS.

Figure 3
Effect of ESM 50 μg/mL on murine BV-2 cells microglial cells viability. Induction control (Cinduction - DMEM with 2% FBS and LPS) and negative control (C- - DMEM with 2% FBS) were used. The test was performed in five independent experiments, each in triplicate. Results were expressed in inhibition percentage compared to the induction control. The mean ± S.E.M. of was used.
Figure 4
Effect of ESM 50 μg/mL on NO production in an in vitro model of LPS-induced neuroinflammation. Murine BV-2 cells microglial cell line was used. Induction control (Cinduction - DMEM with 2% FBS and LPS) and negative control (C- - DMEM with 2% FBS) were used. The test was performed in five independent experiments, each in triplicate. Results were expressed in inhibition percentage compared to the induction control (Cinduction). The mean ± S.E.M. was used. Bars with **** are extremely different (p <0.0001).

In vivo anti-inflammatory and antinociceptive activities

As shown in Figure 5, ESM significantly reduced the mice ear inflammatory edema induced by croton oil at all tested doses. No statistical difference was found when compared to indomethacin, an NSAID often used in clinical practice (Rang et al. 2016RANG HP, DALE MM & RITTER JM. 2016. Farmacologia, 8th ed., Rio de Janeiro: Elsevier, 1939 p. ISBN 9788535283433.). This result is quite interesting as it reinforces the ethnopharmacological uses of S. malaccense.

Figure 5
Topical effect of different doses of ESM on croton-oil-induced ear edema in mice. The vehicle, indomethacin 10 mg/kg, and ESM 50, 100, and 300 mg/kg were administered orally 60 min before topical application of 2.5% croton oil diluted in acetone. The weight difference between the right (inflamed) and the left (non-inflamed) ear biopsies was used to measure edema and, consequently, the inflammatory process. Each column’s values represent the mean±S.E.M. ANOVA followed by the Tukey test, used as post hoc. Significant values:**p < 0.01 and ****p <0.0001 compared to the vehicle group; # means statistically equal to the reference drug (indomethacin).

As shown in Figure 6, ESM reduced the nociceptive stimulus induced by acetic acid in an inverse manner to the dose administered. In addition, Figure 7(a-b) reports that ESM reduced the licking paw time in both formalin test phases. Only the doses of 100 and 300 mg/kg were useful in the neurogenic phase. ESM at all tested doses significantly reduced the nociception in the inflammatory phase; however, the effectiveness of ESM 300 mg/kg was particularly notable.

Figure 6
Effect of different doses of ESM on the number of writhings in acetic acid-induced in mice. The vehicle, indomethacin 10 mg/kg, and ESM 50, 100, and 300 mg/kg were administered orally 60 min before acetic acid 0.6% injection. The number of writhings was counted for 30 min. The values of each column represent the mean±S.E.M. ANOVA followed by the Tukey test, used as post hoc. Significant values:**p < 0.01 and ****p <0.0001 compared to the vehicle group; # means statistically equal to the reference drug (indomethacin).
Figure 7
Effect of different doses of ESM on licking paw time in the first (a) and second (b) phases of the formalin test in mice. Vehicle, indomethacin 10 mg/kg and ESM 50, 100, and 300 mg/kg were administered orally, and morphine 7.5 mg/kg was administered intraperitoneally 60 and 30 min before formalin injection, respectively. The licking paw time was counted for 5 min (first phase) and during 15-30 min (second phase) after formalin injection. Each column’s values represent the mean±S.E.M. ANOVA followed by the Tukey test, used as post hoc. Significant values: *p < 0.05 and ****p <0.0001 compared to the vehicle group.

Chemical characterization of ESM by UHPLC-MS

The public repository of mass spectra for small chemical compounds MassBank (Horai et al. 2010HORAI H ET AL. 2010. MassBank: a public repository for sharing mass spectral data for life sciences. J Mass Spectrom 45: 703-714. https://doi.org/10.1002/jms.1777.) was used as the database to identify the compounds present in ESM. According to the exact mass and the fragments (MS1 and MS2) obtained, three flavonoids: myricitrin (m/z 465.1022) and its aglycone myricetin (m/z 319.0450), quercetin (m/z 303.0504), and the alkaloid reserpine (m/z 609.2713) (Figure 8a-d) were identified.

Figure 8
Chemical structure of myricitrin (a), myricetin (b), quercetin (c), and reserpine (d).

The flavonols aglycones presented predominantly the MS2 fragment m/z 153.0199 and, in accordance to the mass spectrometry fragmentation pathways described (Bindu et al. 2014BINDU S, RAMESHKUMAR KB, KUMAR B, SINGH A & ANILKUMARA C. 2014. Distribution of reserpine in Rauvolfia species from India–HPTLC and LC–MS studies. Ind Crop Prod 62: 430-436. http://dx.doi.org/10.1016/j.indcrop.2014.09.018.), reserpine presented the following MS2 fragments: m/z 448.1910, m/z 397.2070, m/z 365.1790, m/z 195.0630 and m/z 174.0890.

DISCUSSION

Antioxidant activity

Antioxidant effectiveness is related to various distinct aspects, including sample scavenging capacity, concentration, solubility, metal chelating activity, reducing potential, and lipid peroxidation inhibition. Thus, different assays were accomplished (Alam et al. 2013ALAM MN, BRISTI NJ & RAFIQUZZAMAN M. 2013. Review on in vivo and in vitro methods evaluation of antioxidant activity. S P J 21: 143-152. https://doi.org/10.1016/j.jsps.2012.05.002., Scio et al. 2012SCIO E, MENDES RF, MOTTA EVS, BELLOZI PMQ, ARAGÃO DMO, MELLO J, FABRI RL, MOREIRA JR, ASSIS IVL & BOUZADA MLM. 2012. Antimicrobial and Antioxidant Activities of Some Plant Extracts, In: Rao V (Ed), Phytochemicals as Nutraceuticals – Global Approaches to Their Role in Nutrition and Health, p. 21-42. ISBN 978-953-51-0203-8.). DPPH assay was performed as it is quite simple, reproducible, and appropriate to evaluate free radical scavenging activity of compounds with different polarities (Scio et al. 2012SCIO E, MENDES RF, MOTTA EVS, BELLOZI PMQ, ARAGÃO DMO, MELLO J, FABRI RL, MOREIRA JR, ASSIS IVL & BOUZADA MLM. 2012. Antimicrobial and Antioxidant Activities of Some Plant Extracts, In: Rao V (Ed), Phytochemicals as Nutraceuticals – Global Approaches to Their Role in Nutrition and Health, p. 21-42. ISBN 978-953-51-0203-8.). In contrast, β-carotene/linoleic acid bleaching assay is useful to verify the sample ability to inhibit lipid peroxidation, mainly due to its high correlation with the in vivo impairment associated with oxidative stress (Ismail et al. 2004ISMAIL A, MARJAN ZM & FOONG CW. 2004. Total antioxidant activity and phenolic content in selected vegetables. Food Chem 87: 581-586. https://doi.org/10.1016/j.foodchem.2004.01.010.). Also, phosphomolybdenum assay is convenient to verify the total antioxidant capacity of polar and non-polar compounds (Prieto et al. 1999PRIETO P, PINEDA M & AGUILAR M. 1999. Spectrophotometric quantitation of antioxidant capacity through the formation of a phosphomolybdenum complex: specific application to the determination of vitamin E. Anal Biochem 269: 337-341. https://doi.org/10.1006/abio.1999.4019.). The FRAP test is suitable for reducing power assessment (Oyaizu 1986OYAIZU M. 1986. Studies on product of browning reaction prepared from glucose amine. Jap J Nut Diet 44: 307-315. https://doi.org/10.5264/eiyogakuzashi.44.307.).

In summary, the results suggested that ESM presents relevant antioxidant activity (Table I), as IC50 or EC50 values below 100 μg/mL are considered significant for plant extracts, which are endowed with several active and non-active chemical constituents at low concentrations (Cos et al. 2006COS P, VLIETINCK AJ, BERGHE DV & MAES L. 2006. Anti-infective potential of natural products: how to develop a stronger in vitro ‘proof-of-concept’. J Ethnopharmacol 106: 290-302. https://doi.org/10.1016/j.jep.2006.04.003.).

Malonaldehyde (MDA) produced by lipid peroxidation in the TBARs assay is generated in ground beef throughout the experiment (Embuscado 2015EMBUSCADO ME. 2015. Spices and herbs: natural sources of antioxidants – a mini review. J Funct Foods 18: 811-819. https://doi.org/10.1016/j.jff.2015.03.005.). Thus, MDA quantification provides valuable data to predict the in vivo antioxidant potential of ESM.

Antiglycant activity

Glycation is a non-enzymatic reaction in which a reducing sugar binds to a circulating or structural protein, including collagen, neural proteins, hemoglobin, and albumin (Ma et al. 2016MA H, LIU W, FROST L, KIRSCHENBAUM LJ, DAIN JA & SEERAM NP. 2016. Glucitol-core containing gallotannins inhibit the formation of advanced glycation end-products mediated by their antioxidant potential. Food Funct 7: 2213-2222. https://doi.org/10.1039/c6fo00169f.). Advanced Glycation End-products (AGEs) are irreversible heterogeneous compounds produced after subsequent dehydration, oxidation, and cyclization reactions (Grzegorczyk-Karolak et al. 2016GRZEGORCZYK-KAROLAK I, GOŁĄB K, GBUREK J, WYSOKIŃSKA H & MATKOWSKI A. 2016. Inhibition of advanced glycation end-product formation and antioxidant activity by extracts and polyphenols from Scutellaria alpina L. and S. altissima L. Molecules 21: 739. https://doi.org/10.3390/molecules21060739.). AGEs interact with specific receptors (RAGEs) in a variety of cells, which favors the synthesis of reactive oxygen species due to NADPH oxidase activation, and stimulates the NF-κB pathway, inducing the release of several cytokines, including IL-1, IL-6, TNF-α, endothelin-1, and tissue factor, generating an inflammatory process, which becomes the molecular basis of several pathologies, such as diabetes, nephropathies and cardiovascular and neurodegenerative diseases (Chuah et al. 2013CHUAH YK, BASIR R, TALIB H, TIE TH & NORDIN N. 2013. Receptor for advanced glycation end products and its involvement in inflammatory diseases. J Inflamm Res 2013: 403460. http://dx.doi.org/10.1155/2013/403460., Park et al. 2012PARK CH, TANAKA T, KIM HY, PARK JC & YAKOZAWA T. 2012. Protective effects of Corni fructus against advanced glycation end products and radical scavenging. Evid Based Complement Alternat Med 2012: 418953. https://doi.org/ 10.1155/2012/418953., Sun et al. 2016SUN J, LIU W, MA H, MARAIS JPJ, KHOO C, DAIN JA, ROWLEY DC & SEERAM NP. 2016. Effect of cranberry (Vaccinium macrocarpon) oligosaccharides on the formation of advanced glycationend-products. J Berry Res 6: 149-158. https://doi.org/10.3233/JBR-160126.). It is well known that cerebrospinal fluid from patients with Alzheimer’s disease and the frontal cortex of patients with Parkinson present high levels of AGEs (Chen et al. 2012CHEN X, GUO C & KONG J. 2012. Oxidative stress in neurodegenerative diseases. Neural Regen. Res 7: 376-385. https://doi.org/10.3969/j.issn.1673-5374.2012.05.009., Schinkovitz et al. 2018SCHINKOVITZ A ET AL. 2018. Secondary metabolites from lichen as potent inhibitors of advanced glycation end products and vasodilative agents. Fitoterapia 131: 182-188. https://doi.org/ 10.1016/j.fitote.2018.10.015.).

The antiglycant effect of ESM may be attributed, at least in part, to its antioxidant capacity, as reactive oxygen species are generated during all the steps of glycation reaction (Liu et al. 2018LIU J, HE Y, WANG S, HE Y, WANG W, LI Q & CAO X. 2018. Ferulic acid inhibits advanced glycation end products (AGEs) formation and mitigates the AGEs-induced inflammatory response in HUVEC cells. J Funct Foods 48: 19-26. https://doi.org/10.1016/j.jff.2018.06.024.). The antioxidant and antiglycation effectiveness of ESM encouraged evaluating its activity after LPS-induced inflammation in microglial cells. This assay is mediated by nitric oxide free radical and may be useful to predict ESM anti-neuroinflammatory potential.

In vitro evaluation of the anti-neuroinflammatory activity of ESM in BV-2 microglial cell line

Neuroinflammation plays an important role in the pathogenesis of several neurodegenerative diseases. Microglia cells are brain-resident phagocytes that exert various functions related to the host defense when activated (Bozic et al. 2015BOZIC I, SAVIC D, LAKETA D, BJELOBABA I, MILENKOVIC I, PEKOVIC S, NEDELJKOVIC N & LAVRNJA I. 2015. Benfotiamine attenuates inflammatory response in LPS stimulated BV-2 microglia. PLoS ONE 10: e0118372. https://doi.org/10.1371/journal.pone.0118372.). Microglia activation is characterized by the release of different pro-inflammatory mediators, such as interleukins, tumor necrosis factor-alpha (TNF-α), and reactive oxygen species (ROS), including nitric oxide (NO). These mechanisms may be induced by exposure to toxins, including LPS, and are strictly related to neuroinflammation and brain injuries (Gan et al. 2015GAN P ET AL. 2015. Anti-inflammatory effects of glaucocalyxin B in microglia cells. J Pharmacol Sci 128: 35-46. https://doi.org/10.1016/j.jphs.2015.04.005.).

Although NO is an important biologically active molecule which fulfills several functions in the body, its high levels, produced by activated microglial cells, may induce the formation of reactive oxygen species, which generates brain oxidative stress and contributes to the neuronal cell death and the oligodendrocyte degeneration process associated with demyelinating diseases (Eguchi et al. 2011EGUCHI H, FUJIWARA N, SAKIYAMA H, YOSHIHARA D & SUZUKI K. 2011. Hydrogen peroxide enhances LPS-induced nitric oxide production via the expression of interferon beta in BV-2 microglial cells. Neurosci Lett 494: 29-33. https://doi.org/10.1016/j.neulet.2011.02.047.).

Several different mechanisms can inhibit the increase of NO levels by LPS exposure; however, ESM might reduce them by the neutralization of the NO• radical itself. More importantly, those results, together with the inhibition of AGEs formation, suggested that ESM presented neuroprotective potential, which is quite relevant due to the current emerging of neurodegenerative diseases (Elmann et al. 2011ELMANN A, MORDECHAY S, ERLANK H, TELERMAN A, RINDNER M & OFIR R. 2011. Anti-neuroinflammatory effects of the extract of Achillea fragrantissima A. BMC Complement Altern Med 11: 1198. https://doi.org/10.1186/1472-6882-11-98., Kim et al. 2018KIM S, BAEK SK & SONG KB. 2018. Physical and antioxidant properties of alginate films prepared from Sargassum fulvellum with black chokeberry extract. Food Packag Shelf Life 18: 157-163. https://doi.org/10.1186/1472-6882-11-98., Zhang et al. 2010ZHANG C, BROWNE A, CHILD D, DIVITO JR, STEVENSON JA & TANZI RE. 2010. Loss of function of ATXN1 increases amyloid beta-protein levels by potentiating beta-secretase processing of beta-amyloid precursor protein. J Biol Chem 285: 8515-8526. https://doi.org/10.1074/jbc.M109.079079.).

In vivo anti-inflammatory and antinociceptive activities

Croton oil-induced ear edema test

Croton oil is a natural product endowed with phorbol esters, which stimulate the release of several inflammatory mediators, including transcription factors, cytokines, and enzymes, such as cyclooxygenase, 5-lipoxygenase, and phospholipase-A2 (Pascual & Glass 2006PASCUAL G & GLASS CK. 2006. Nuclear receptor versus inflammation: mechanisms of transrepression. Trends Endocrinol. Metab 17: 321-327 http://dx.doi.org/10.1016/j.tem.2006.08.005., Saraiva et al. 2011SARAIVA RA ET AL. 2011. Topical anti-inflammatory effect of Caryocar coriaceum Wittm. (Caryocaraceae) fruit pulp fixed oil on mice ear edema induced by different irritant agents. J Ethnopharmacol 136: 504-510. http://dx.doi.org/10.1016/j.jep.2010.07.002.). For this reason, different inflammatory pathways are activated, so that croton oil-induced ear edema test is entirely appropriated to verify a possible anti-inflammatory activity of a tested drug independent of its mechanism of action (Pinto et al. 2015PINTO NCC, MACHADO DC, SILVA JM, CONEGUNDES JLM, GUALBERTO ACM, GAMEIRO J, CHEDIER LM, CASTAÑON MCMN & SCIO E. 2015. Pereskia aculeata Miller leaves present in vivo topical anti-inflammatory activity in models of acute and chronic dermatitis. J Ethnopharmacol 173: 330-337. https://doi.org/10.1016/j.jep.2015.07.032.).

Although the phlogistic agent was topically applied in this test, this screening assay was used to verify the action of anti-inflammatory chemical compounds administered both topically and orally. This test is also entirely appropriated to evaluate natural compounds, as it requires small amounts, and it is rapidly accomplished with reproducible results (Gábor 2003GÁBOR M. 2003. Models of acute inflammation in the ear. In: Winyard PG & Willoughby DA (Eds), Inflammation Protocols, New Jersey: Humana Press, New Jersey, USA, p. 129-137. https://doi.org/10.1385/1-59259-374-7:129.).

Although many mediators are involved in the inflammatory processes induced by croton oil (Pascual & Glass 2006PASCUAL G & GLASS CK. 2006. Nuclear receptor versus inflammation: mechanisms of transrepression. Trends Endocrinol. Metab 17: 321-327 http://dx.doi.org/10.1016/j.tem.2006.08.005., Saraiva et al. 2011SARAIVA RA ET AL. 2011. Topical anti-inflammatory effect of Caryocar coriaceum Wittm. (Caryocaraceae) fruit pulp fixed oil on mice ear edema induced by different irritant agents. J Ethnopharmacol 136: 504-510. http://dx.doi.org/10.1016/j.jep.2010.07.002.), it was not possible to predict ESM mechanism of action. However, its antioxidant capacity may contribute, at least in part, to reduce the mice ear edema. It is well known that oxidative stress is associated with inflammation development (Bhagavan et al. 2013BHAGAVAN NB, ARUNACHALAM S, DHASARATHAN P & KANNAN ND. 2013. Evaluation of anti-inflammatory activity of Indigofera aspalathoides Vahl in Swiss albino mice. J Pharm Res 6: 350-354. https://doi.org/10.1016/j.jopr.2013.02.018.).

Acetic acid-induced writhing test

Both central analgesics and anti-inflammatory agents are suitable to respond to this test. The intraperitoneal injection of acetic acid in mouse induces the synthesis of eicosanoids and other inflammatory mediators, such as histamine and bradykinin, and the release of several cytokines, including TNF-α, IL-1, IL-6, and IL-8, which irritate the serous abdominal membranes and stimulate nociceptive neurons (Favero et al. 2014FAVERO FF, GRANDO R, NONATO FR, SOUSA IMM, QUEIROZ NCA, LONGATO GB, ZAFRED RRT, CARVALHO JE, SPINDOLA HM & FOGLIO MA. 2014. Artemisia annua L.: evidence of sesquiterpene lactones’ fraction antinociceptive activity. BMC Complement Altern 14: 266. http://dx.doi.org/10.1186/1472-6882-14-266., Pinheiro et al. 2011PINHEIRO B, SILVA ASB, SOUZA GEP, FIGUEIREDO JG, CUNHA FQ, LAHLOU S, SILVA JKR, MAIA JGS & SOUSA PJC. 2011. Chemical composition, antinociceptive and anti-inflammatory effects in rodents of the essential oil of Peperomia serpens (Sw.) Loud. J Ethnopharmacol 188: 479-486. https://doi.org/10.1016/j.jep.2011.09.037.). Mice respond to this test with stereotyped movements, characterized by abdominal writhing, dorsal abdominal muscle sprain and contraction of the whole body (Silva et al. 2013SILVA VG ET AL. 2013. Anti-inflammatory and Antinociceptive Activity of Epiisopiloturine, an Imidazole Alkaloid Isolated from Pilocarpus microphyllus. J Nat Prod 76: 1071-1077. 10.1021/np400099m.).

As shown in Figure 6, ESM reduced the nociceptive stimulus induced by acetic acid in an inverse manner to the dose administered. This find was not surprising, as similar dose-response curves of plant extracts in in vivo nociceptive models have been reported in the literature (Pinto et al. 2015PINTO NCC, MACHADO DC, SILVA JM, CONEGUNDES JLM, GUALBERTO ACM, GAMEIRO J, CHEDIER LM, CASTAÑON MCMN & SCIO E. 2015. Pereskia aculeata Miller leaves present in vivo topical anti-inflammatory activity in models of acute and chronic dermatitis. J Ethnopharmacol 173: 330-337. https://doi.org/10.1016/j.jep.2015.07.032., Huerta-Reyes et al. 2013HUERTA-REYES M, HERRERA-RUIZ M, GONZÁLEZ-CORTAZAR M, ZAMILPA A, LEÓN E, REYES-CHILPA R, AGUILAR-ROJAS A & TORTORIELLO J. 2013. Neuropharmacological in vivo effects and phytochemical profile of the extract from the aerial parts of Heteropterys brachiata (L.) DC. (Malpighiaceae). J Ethnopharmacol 146: 311-317. https://doi.org/10.1016/j.jep.2012.12.049.). According to Williamson et al. (1996)WILLIAMSON EM, OKPAKO DT & EVANS FJ. 1996. Selection, Preparation and Pharmacological Evaluation of Plant Material, Chichester: J Wiley & Sons, 238 p. ISBN: 978-0-471-94217-7. it may be explained by the presence of several compounds in plant extracts, as a competitive or non-competitive antagonism may occur. An agonist’s threshold dose may not be significantly high; however, in non-competitive antagonism, the maximum response may be decreased by an antagonist compound, which may be active even beyond the receptor, interfering in intracellular second messengers signaling pathways.

Formalin-induced paw licking test

This test helps distinguish whether a tested drug can inhibit nociception stimulus by central or peripheral mechanisms. The sub-plantar injection of formalin induces a biphasic behavioral response characterized by paw licking. In the first (neurogenic) phase, up to 5 min after the formalin injection, there is a direct sensitization of C and Aδ fibers due to the release of neurogenic mediators, including excitatory amino acids, nitric oxide, and substance P. The second (inflammatory) phase, which occurs between 15 and 30 min after formalin injection, is related to the release of several inflammatory mediators, such as prostaglandins, histamine, bradykinin, and serotonin (Hunskaar et al. 1985HUNSKAAR S, FASMER OB & HOLE K. 1985. The formalin test in mice, a useful technique for evaluating mild analgesia. J Neurosci Methods 14: 69-76. https://doi.org/10.1016/0165-0270(85)90116-5., Saldanha et al. 2017SALDANHA AA, SIQUEIRA JM, CASTRO AHF, MATOS NA, KLEIN A, SILVA DB, CAROLLO CA & SOARES AC. 2017. Peripheral and central antinociceptive effects of the butanolic fraction of Byrsonima verbascifolia leaves on nociception-induced models in mice. Inflammopharmacology 25: 81-90. https://doi.org/10.1007/s10787-016-0300-5., Silva et al. 2013SILVA VG ET AL. 2013. Anti-inflammatory and Antinociceptive Activity of Epiisopiloturine, an Imidazole Alkaloid Isolated from Pilocarpus microphyllus. J Nat Prod 76: 1071-1077. 10.1021/np400099m.). Thus, antinociceptive drugs that act centrally or in peripheral fibers, interfering in the central nociceptive pathways, are capable of inhibiting the licking paw time of both phases. Simultaneously, those that interfere in peripheral mechanisms can reduce the licking paw time only in the second phase (Monteiro et al. 2014MONTEIRO EMH ET AL. 2014. Antinociceptive and anti-inflammatory activities of the sesame oil and sesamin. Nutrients 6: 1931-1944. https://doi.org/10.3390/nu6051931.). For those reasons, the opioid morphine and the NSAID indomethacin were both used as reference drugs.

Central non-opioid analgesics are active in both phases; however, they are more efficacious in the second phase (Valerio et al. 2009VALERIO DA ET AL. 2009. Quercetin reduces inflammatory pain: inhibition of oxidative stress and cytokine production. J Nat Prod 72: 1975-1979. https://doi.org/10.1021/np900259y.), explaining the mice’s response towards ESM 300 mg/kg. It is noteworthy that ESM is a crude extract endowed with several chemical constituents acting separately as central and peripheral analgesics in synergism.

The chemical compounds produced during the oxidative stress, including hydrogen peroxide, peroxynitrite, and superoxide anion, are important mediators in inflammatory pain and may cause tissue injuries. Thus, antioxidant agents may reduce the nociceptive stimulus preventing the formation of free radicals in the site of inflammation or even centrally in the spinal cord (Valerio et al. 2009VALERIO DA ET AL. 2009. Quercetin reduces inflammatory pain: inhibition of oxidative stress and cytokine production. J Nat Prod 72: 1975-1979. https://doi.org/10.1021/np900259y.). As ESM showed remarkable antioxidant capacity, it is reasonable to speculate that the inhibition of free radicals may explain, at least in part, its antinociceptive activity.

Chemical compounds identified in ESM by UHPLC-MS

The anti-inflammatory and antioxidant activities of myricitrin are well known. Its ability to reduce the neuroinflammatory process was recently reported, which involves decreasing several chemical mediators, including NF-κB and MAPK signaling pathways, IL-1β, IL-6, TNF-α, MCP-1, and the enzymes COX-2 and iNOS (Yang et al. 2019YANG YL, LIU M, CHENG X, LI W, ZHANG S, WANG Y & DU G. 2019. Myricitrin blocks activation of NF-κB and MAPK signaling pathways to protect nigrostriatum neuron in LPS-stimulated mice. J Neuroimmunol 337: 577049. https://doi.org/10.1016/j.jneuroim.2019.577049.). Also, Sobeh et al. (2019)SOBEH M, PETRUK G, OSMAN S, EL-RAEY MA, IMBIMBO P, MONTI DM & WINK M. 2019. Isolation of Myricitrin and 3,5-di-O-Methyl Gossypetin from Syzygium samarangense and Evaluation of their Involvement in Protecting Keratinocytes against Oxidative Stress via Activation of the Nrf-2 Pathway. Molecules 24: 1839. https://doi.org/10.3390/molecules24091839. demonstrated that myricitrin significantly modified ROS generation profile, glutathione levels and protein oxidation by interfering with mitogen-activated protein kinase (MAPK) signaling pathways, using the sodium arsenite-induced oxidative stress model on human keratinocytes. Besides, myricitrin presents relevant potential as a natural agent to treat neurodegenerative disorders, as this flavonoid can suppress the aggregation of various aberrant proteins, eliminate several abnormal proteins from cell environment and reduce the inclusions of misfolded proteins, which decreases the neurotoxicity of these anomalous biomolecules (Joshi et al. 2019JOSHI V, MISHRA R, UPADHYAY A, AMANULLAH A, POLURI KM, SINGH S, KUMAR A & MISHRA A. 2019. Polyphenolic flavonoid (Myricetin) upregulated proteasomal degradation mechanisms: Eliminates neurodegenerative proteins aggregation. J Cell Physiol 234: 20900-20914. https://doi.org/10.1002/jcp.28695.). It is also noteworthy to mention that a previous study reported that four flavan-3-ol derivatives, including (+) – catechin, mearnsitrin, myricitrin, and quercitrin isolated from S. malaccense leaves exhibit inhibitory activity against COX-1 and COX-2 enzymes (Noreen et al. 1998NOREEN Y, SERRANO G, PERERA P & BOHLIN L. 1998. Flavan-3-ols isolated from some medicinal plants inhibiting COX-1 and COX-2 catalysed prostaglandin biosynthesis. Planta Med 64: 520-524. https://doi.org/10.1055/s-2006-957506.). Besides, Arumugam et al. (2019)ARUMUGAM B, PALANISAMY UD, CHUA KH & KUPPUSAMY UR. 2019. Protective effect of myricetin derivatives from Syzygium malaccense against hydrogen peroxide-induced stress in ARPE-19 cells. Mol Vis 25: 47-59. http://www.molvis.org/molvis/v25/47. showed that a myricetin enriched fraction obtained from S malaccense inhibited the oxidative stress induced by hydrogen peroxide in pigmented epithelial cells of the human retina (ARPE-19).

The review carried out by Calis et al. (2020)CALIS Z, MOGULKOC R & BALTACI AK. 2020. The roles of flavonoles/flavonoids in neurodegeneration and neuroinflammation. Mini Rev Med Chem 20: 1475-1488. https://doi.org/10.2174/1389557519666190617150051. pointed that a variety of flavonoids, especially those obtained from diet, including quercetin, have received great attention from the scientific community due to their potential to prevent neuroinflammation and neurodegenerative process. On the other hand, reserpine is recognized as a classical antipsychotic and antihypertensive drug used in clinical practice. However, studies have reported that reserpine presents neurotoxicity in zebrafish (Danio rerio) assays (Wang et al. 2019WANG S ET AL. 2019. Developmental neurotoxicity of reserpine exposure in zebrafish larvae (Danio rerio). Comparative Biochemistry and Physiology Part C: Regul Toxicol Pharmacol 223: 115-123. https://doi.org/10.1016/j.cbpc.2019.05.008.) and may induce the development of depression and Parkinson’s disease Khan et al. 2018KHAN ZA, SHAHZAD SA, ANJUM A, BALE AT & NAQVI SAR. 2018. Synthetic approaches toward the reserpine. Synth Commun 48: 1128-1147. https://doi.org/10.1080/00397911.2018.1434546., Rijntjes & Meyer 2019RIJNTJES M & MEYER PT. 2019. No free lunch with herbal preparations: lessons from a case of parkinsonism and depression due to herbal medicine containing reserpine. Front Neurol 10: 634. https://doi.org/ 10.3389/fneur.2019.00634.).

The flavonoids myricitrin, myricetin and quercetin, have been identified in fruits and leaves of S. malaccense (Arumugam et al. 2016ARUMUGAM B, MANAHARAN T, HENG CK, KUPPUSAMY UR & PALANISAMY UD. 2016. Antioxidant and antiglycemic potentials of a standardized extract of Syzygium malaccense. LWT - Food Sci Technol 59: 707-712. https://doi.org/10.1016/j.lwt.2014.06.041., 2019, Batista 2017BATISTA AG. 2017. Red-jambo (Syzygium malaccense): bioactive compounds in fruits and leaves. LWT - Food Sci Technol 76: 284-291. https://doi.org/10.1016/j.lwt.2016.05.013.). However, no studies were reporting the identification of the alkaloid reserpine in this plant species as far as we know. To confirm this alkaloid’s presence, the standard reserpine and ESM were eluted and coeluted at the same analytical conditions. The signals related to reserpine acquired after the standard, ESM, and co-elution injections, showed similar UV profile (UV max 217, 268 and 299 nm) and retention time (RT = 18.4 min), confirming the reserpine identification obtained by the mass spectrum analysis.

CONCLUSION

The present study corroborates to the ethnopharmacological uses of S. malaccense, as the methanolic extract obtained from the leaves showed relevant in vivo anti-inflammatory and antinociceptive effects. Besides, the anti-inflammatory action of S. malaccense also occurred in vitro on BV-2 microglial cells after LPS addition, which revealed the ESM potential to be used as a neuroprotective agent. The antioxidant and antiglycant activities may be related, at least in part, to the mechanism of the in vitro neuroinflammation inhibition promoted by ESM. This result is quite important due to the role of brain inflammation in the pathogenesis of neurodegenerative disorders. Although reserpine was identified in ESM, the flavonoids myricitrin, myricetin and quercetin, among other compounds, are more likely to be responsible for those effects.

ACKNOWLEDGMENTS

This work was supported by Coordenação de Aperfeiçoamento de Pessoal de Nível Superior CAPES/PDSE [CEX APQ 01403-14]; and Fundação de Amparo à Pesquisa do Estado de Minas Gerais FAPEMIG (APQ 00487-16). The authors are grateful to UMinho for the opportunity for academic-scientific exchange, to the Reproduction Biology Center of the Universidade Federal de Juiz de Fora for providing the mice, to the Laboratory of Chemistry of Bioactive Natural Products, René Rachou Research Center for the UHPLC-MS data, and to Delfino Antônio Campos for technical assistance.

REFERENCES

  • ADEBAYO AH, OGUNDARE OC & ADEGBITE OS. 2015. Sub-acute evaluation of extract of Syzygium malaccense in albino rats. J Med Plant Res 9: 60-71. https://doi.org/10.3923/rjmp.2015.60.71.
  • ALAM MN, BRISTI NJ & RAFIQUZZAMAN M. 2013. Review on in vivo and in vitro methods evaluation of antioxidant activity. S P J 21: 143-152. https://doi.org/10.1016/j.jsps.2012.05.002.
  • ARUMUGAM B, MANAHARAN T, HENG CK, KUPPUSAMY UR & PALANISAMY UD. 2016. Antioxidant and antiglycemic potentials of a standardized extract of Syzygium malaccense. LWT - Food Sci Technol 59: 707-712. https://doi.org/10.1016/j.lwt.2014.06.041.
  • ARUMUGAM B, PALANISAMY UD, CHUA KH & KUPPUSAMY UR. 2019. Protective effect of myricetin derivatives from Syzygium malaccense against hydrogen peroxide-induced stress in ARPE-19 cells. Mol Vis 25: 47-59. http://www.molvis.org/molvis/v25/47.
  • BATISTA AG. 2017. Red-jambo (Syzygium malaccense): bioactive compounds in fruits and leaves. LWT - Food Sci Technol 76: 284-291. https://doi.org/10.1016/j.lwt.2016.05.013.
  • BHAGAVAN NB, ARUNACHALAM S, DHASARATHAN P & KANNAN ND. 2013. Evaluation of anti-inflammatory activity of Indigofera aspalathoides Vahl in Swiss albino mice. J Pharm Res 6: 350-354. https://doi.org/10.1016/j.jopr.2013.02.018.
  • BINDU S, RAMESHKUMAR KB, KUMAR B, SINGH A & ANILKUMARA C. 2014. Distribution of reserpine in Rauvolfia species from India–HPTLC and LC–MS studies. Ind Crop Prod 62: 430-436. http://dx.doi.org/10.1016/j.indcrop.2014.09.018.
  • BLOIS MS. 1958. Antioxidant determination by the use of stable free radical. Nature 181: 1199-2000. https://doi.org/10.1038/1811199a0.
  • BOZIC I, SAVIC D, LAKETA D, BJELOBABA I, MILENKOVIC I, PEKOVIC S, NEDELJKOVIC N & LAVRNJA I. 2015. Benfotiamine attenuates inflammatory response in LPS stimulated BV-2 microglia. PLoS ONE 10: e0118372. https://doi.org/10.1371/journal.pone.0118372.
  • CALIS Z, MOGULKOC R & BALTACI AK. 2020. The roles of flavonoles/flavonoids in neurodegeneration and neuroinflammation. Mini Rev Med Chem 20: 1475-1488. https://doi.org/10.2174/1389557519666190617150051.
  • CAMPBELL G, DARKE S, BRUNO R & DEGENHARDT L. 2015. The prevalence and correlates of chronic pain and suicidality in a nationally representative sample. Aust N Z J Psychiatry 49: 803-811. https://doi.org/10.1177/0004867415569795.
  • CHEN X, GUO C & KONG J. 2012. Oxidative stress in neurodegenerative diseases. Neural Regen. Res 7: 376-385. https://doi.org/10.3969/j.issn.1673-5374.2012.05.009.
  • CHUAH YK, BASIR R, TALIB H, TIE TH & NORDIN N. 2013. Receptor for advanced glycation end products and its involvement in inflammatory diseases. J Inflamm Res 2013: 403460. http://dx.doi.org/10.1155/2013/403460.
  • COS P, VLIETINCK AJ, BERGHE DV & MAES L. 2006. Anti-infective potential of natural products: how to develop a stronger in vitro ‘proof-of-concept’. J Ethnopharmacol 106: 290-302. https://doi.org/10.1016/j.jep.2006.04.003.
  • DUNSTAN CA. 1997. Evaluation of some Samoan and Peruvian medicinal plants by prostaglandin biosynthesis and rat ear oedema assays. J Ethnopharmacol 57: 35-56. https://doi.org/10.1016/s0378-8741(97)00043-3.
  • EGUCHI H, FUJIWARA N, SAKIYAMA H, YOSHIHARA D & SUZUKI K. 2011. Hydrogen peroxide enhances LPS-induced nitric oxide production via the expression of interferon beta in BV-2 microglial cells. Neurosci Lett 494: 29-33. https://doi.org/10.1016/j.neulet.2011.02.047.
  • ELMANN A, MORDECHAY S, ERLANK H, TELERMAN A, RINDNER M & OFIR R. 2011. Anti-neuroinflammatory effects of the extract of Achillea fragrantissima A. BMC Complement Altern Med 11: 1198. https://doi.org/10.1186/1472-6882-11-98.
  • EMBUSCADO ME. 2015. Spices and herbs: natural sources of antioxidants – a mini review. J Funct Foods 18: 811-819. https://doi.org/10.1016/j.jff.2015.03.005.
  • FARSI DA, HARRIS CS, REID L, BENNET SAL, HADDAD PS, MARTINEAU LC & ARNASON JT. 2008. Inhibition of non-enzymatic glycation by silk extracts from a Mexican land race and modern inbred lines of maize (Zea mays). Phytother Res 22: 108-112. https://doi.org/10.1002/ptr.2275.
  • FAVERO FF, GRANDO R, NONATO FR, SOUSA IMM, QUEIROZ NCA, LONGATO GB, ZAFRED RRT, CARVALHO JE, SPINDOLA HM & FOGLIO MA. 2014. Artemisia annua L.: evidence of sesquiterpene lactones’ fraction antinociceptive activity. BMC Complement Altern 14: 266. http://dx.doi.org/10.1186/1472-6882-14-266.
  • FAYAZ A, CROFT P, LANGFORD RM, DONALDSON LJ & JONES GT. 2016. Prevalence of chronic pain in the UK: a systematic review and meta-analysis of population studies. BMJ Open 6: e010364. https://doi.org/10.1136/bmjopen-2015-010364.
  • FERNANDES FAN & RODRIGUES S. 2018. Jambo - Syzygium malaccense. Exotic Fruits 245-249. https://doi.org/10.1016/B978-0-12-803138-4.00031-9.
  • GÁBOR M. 2003. Models of acute inflammation in the ear. In: Winyard PG & Willoughby DA (Eds), Inflammation Protocols, New Jersey: Humana Press, New Jersey, USA, p. 129-137. https://doi.org/10.1385/1-59259-374-7:129.
  • GAN P ET AL. 2015. Anti-inflammatory effects of glaucocalyxin B in microglia cells. J Pharmacol Sci 128: 35-46. https://doi.org/10.1016/j.jphs.2015.04.005.
  • GRZEGORCZYK-KAROLAK I, GOŁĄB K, GBUREK J, WYSOKIŃSKA H & MATKOWSKI A. 2016. Inhibition of advanced glycation end-product formation and antioxidant activity by extracts and polyphenols from Scutellaria alpina L. and S. altissima L. Molecules 21: 739. https://doi.org/10.3390/molecules21060739.
  • HORAI H ET AL. 2010. MassBank: a public repository for sharing mass spectral data for life sciences. J Mass Spectrom 45: 703-714. https://doi.org/10.1002/jms.1777.
  • HUERTA-REYES M, HERRERA-RUIZ M, GONZÁLEZ-CORTAZAR M, ZAMILPA A, LEÓN E, REYES-CHILPA R, AGUILAR-ROJAS A & TORTORIELLO J. 2013. Neuropharmacological in vivo effects and phytochemical profile of the extract from the aerial parts of Heteropterys brachiata (L.) DC. (Malpighiaceae). J Ethnopharmacol 146: 311-317. https://doi.org/10.1016/j.jep.2012.12.049.
  • HUNSKAAR S, FASMER OB & HOLE K. 1985. The formalin test in mice, a useful technique for evaluating mild analgesia. J Neurosci Methods 14: 69-76. https://doi.org/10.1016/0165-0270(85)90116-5.
  • ISMAIL A, MARJAN ZM & FOONG CW. 2004. Total antioxidant activity and phenolic content in selected vegetables. Food Chem 87: 581-586. https://doi.org/10.1016/j.foodchem.2004.01.010.
  • JOSHI V, MISHRA R, UPADHYAY A, AMANULLAH A, POLURI KM, SINGH S, KUMAR A & MISHRA A. 2019. Polyphenolic flavonoid (Myricetin) upregulated proteasomal degradation mechanisms: Eliminates neurodegenerative proteins aggregation. J Cell Physiol 234: 20900-20914. https://doi.org/10.1002/jcp.28695.
  • KHAN ZA, SHAHZAD SA, ANJUM A, BALE AT & NAQVI SAR. 2018. Synthetic approaches toward the reserpine. Synth Commun 48: 1128-1147. https://doi.org/10.1080/00397911.2018.1434546.
  • KIM S, BAEK SK & SONG KB. 2018. Physical and antioxidant properties of alginate films prepared from Sargassum fulvellum with black chokeberry extract. Food Packag Shelf Life 18: 157-163. https://doi.org/10.1186/1472-6882-11-98.
  • KOSTER R, ANDERSON M & DE BEER EJ. 1959. Acetic acid for analgesic screening. Fed Proc 18: 412-418.
  • LI J, LIU D, SUN L, LU Y & ZHANG Z. 2012. Advanced glycation end products and neurodegenerative diseases: mechanisms and perspective. J Neurol Sci 317: 1-5. https://doi.org/10.1016/j.jns.2012.02.018.
  • LIU J, HE Y, WANG S, HE Y, WANG W, LI Q & CAO X. 2018. Ferulic acid inhibits advanced glycation end products (AGEs) formation and mitigates the AGEs-induced inflammatory response in HUVEC cells. J Funct Foods 48: 19-26. https://doi.org/10.1016/j.jff.2018.06.024.
  • MA H, LIU W, FROST L, KIRSCHENBAUM LJ, DAIN JA & SEERAM NP. 2016. Glucitol-core containing gallotannins inhibit the formation of advanced glycation end-products mediated by their antioxidant potential. Food Funct 7: 2213-2222. https://doi.org/10.1039/c6fo00169f.
  • MONTEIRO EMH ET AL. 2014. Antinociceptive and anti-inflammatory activities of the sesame oil and sesamin. Nutrients 6: 1931-1944. https://doi.org/10.3390/nu6051931.
  • MOSMANN T. 1983. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods 65: 55-63. https://doi.org/10.1016/0022-1759(83)90303-4.
  • NEAL M & RICHARDSON JR. 2018. Time to get personal: A framework for personalized targeting of oxidative stress in neurotoxicity and neurodegenerative disease. Curr Opin Toxicol 7: 127-132. https://doi.org/10.1016/j.cotox.2018.02.003.
  • NOREEN Y, SERRANO G, PERERA P & BOHLIN L. 1998. Flavan-3-ols isolated from some medicinal plants inhibiting COX-1 and COX-2 catalysed prostaglandin biosynthesis. Planta Med 64: 520-524. https://doi.org/10.1055/s-2006-957506.
  • NUNES PC, AQUINO JS, ROCKENBACH II & STAMFORD TLM. 2016. Physico-chemical characterization, bioactive compounds and antioxidant activity of malay apple [Syzygium malaccense (L.) Merr. & L.M. Perry]. PLoS ONE 11: 1-11. https://doi.org/10.1371/journal.pone.0158134.
  • OYAIZU M. 1986. Studies on product of browning reaction prepared from glucose amine. Jap J Nut Diet 44: 307-315. https://doi.org/10.5264/eiyogakuzashi.44.307.
  • PARK CH, TANAKA T, KIM HY, PARK JC & YAKOZAWA T. 2012. Protective effects of Corni fructus against advanced glycation end products and radical scavenging. Evid Based Complement Alternat Med 2012: 418953. https://doi.org/ 10.1155/2012/418953.
  • PASCUAL G & GLASS CK. 2006. Nuclear receptor versus inflammation: mechanisms of transrepression. Trends Endocrinol. Metab 17: 321-327 http://dx.doi.org/10.1016/j.tem.2006.08.005.
  • PEDROLLO CT, KINUPP VF, SHEPARD-JR G & HEINRICH M. 2016. Medicinal plants at Rio Jauaperi, Brazilian Amazon: Ethnobotanical survey and environmental conservation. J Ethnopharmacol 186: 111-124. https://doi.org/10.1016/j.jep.2016.03.055.
  • PINHEIRO B, SILVA ASB, SOUZA GEP, FIGUEIREDO JG, CUNHA FQ, LAHLOU S, SILVA JKR, MAIA JGS & SOUSA PJC. 2011. Chemical composition, antinociceptive and anti-inflammatory effects in rodents of the essential oil of Peperomia serpens (Sw.) Loud. J Ethnopharmacol 188: 479-486. https://doi.org/10.1016/j.jep.2011.09.037.
  • PINTO NCC, MACHADO DC, SILVA JM, CONEGUNDES JLM, GUALBERTO ACM, GAMEIRO J, CHEDIER LM, CASTAÑON MCMN & SCIO E. 2015. Pereskia aculeata Miller leaves present in vivo topical anti-inflammatory activity in models of acute and chronic dermatitis. J Ethnopharmacol 173: 330-337. https://doi.org/10.1016/j.jep.2015.07.032.
  • PRIETO P, PINEDA M & AGUILAR M. 1999. Spectrophotometric quantitation of antioxidant capacity through the formation of a phosphomolybdenum complex: specific application to the determination of vitamin E. Anal Biochem 269: 337-341. https://doi.org/10.1006/abio.1999.4019.
  • RAMADHANIA ZM, INSANU M, GUNARTI NS, WIRASUTISNA KR, SUKRASNO S & HARTATI R. 2017. Antioxidant activity from ten species of Myrtaceae. Asian J Pharm Clin Res Special Iissue May. https://doi.org/10.22159/ajpcr.2017.v10s2.19470.
  • RANG HP, DALE MM & RITTER JM. 2016. Farmacologia, 8th ed., Rio de Janeiro: Elsevier, 1939 p. ISBN 9788535283433.
  • RIJNTJES M & MEYER PT. 2019. No free lunch with herbal preparations: lessons from a case of parkinsonism and depression due to herbal medicine containing reserpine. Front Neurol 10: 634. https://doi.org/ 10.3389/fneur.2019.00634.
  • SALDANHA AA, SIQUEIRA JM, CASTRO AHF, MATOS NA, KLEIN A, SILVA DB, CAROLLO CA & SOARES AC. 2017. Peripheral and central antinociceptive effects of the butanolic fraction of Byrsonima verbascifolia leaves on nociception-induced models in mice. Inflammopharmacology 25: 81-90. https://doi.org/10.1007/s10787-016-0300-5.
  • SARAIVA RA ET AL. 2011. Topical anti-inflammatory effect of Caryocar coriaceum Wittm. (Caryocaraceae) fruit pulp fixed oil on mice ear edema induced by different irritant agents. J Ethnopharmacol 136: 504-510. http://dx.doi.org/10.1016/j.jep.2010.07.002.
  • SCHIANTARELLI P, CADEL S, ACERBI D & PAVESI L. 1982. Anti-inflammatory activity and bioavailability of percutaneous piroxican. Drug Res 32: 230-235.
  • SCHINKOVITZ A ET AL. 2018. Secondary metabolites from lichen as potent inhibitors of advanced glycation end products and vasodilative agents. Fitoterapia 131: 182-188. https://doi.org/ 10.1016/j.fitote.2018.10.015.
  • SCHMIDT HW & KELM M. 1996. Determination of nitrite and nitrate by the Griess reaction. In: FEELISCH M & STAMLER JS (Eds.), Methods in Nitric Oxide Research, New York: J Wiley & Sons Ltd., New York, USA, p. 491-497.
  • SCIO E, MENDES RF, MOTTA EVS, BELLOZI PMQ, ARAGÃO DMO, MELLO J, FABRI RL, MOREIRA JR, ASSIS IVL & BOUZADA MLM. 2012. Antimicrobial and Antioxidant Activities of Some Plant Extracts, In: Rao V (Ed), Phytochemicals as Nutraceuticals – Global Approaches to Their Role in Nutrition and Health, p. 21-42. ISBN 978-953-51-0203-8.
  • SILVA VG ET AL. 2013. Anti-inflammatory and Antinociceptive Activity of Epiisopiloturine, an Imidazole Alkaloid Isolated from Pilocarpus microphyllus. J Nat Prod 76: 1071-1077. 10.1021/np400099m.
  • SOBEH M, PETRUK G, OSMAN S, EL-RAEY MA, IMBIMBO P, MONTI DM & WINK M. 2019. Isolation of Myricitrin and 3,5-di-O-Methyl Gossypetin from Syzygium samarangense and Evaluation of their Involvement in Protecting Keratinocytes against Oxidative Stress via Activation of the Nrf-2 Pathway. Molecules 24: 1839. https://doi.org/10.3390/molecules24091839.
  • SUN J, LIU W, MA H, MARAIS JPJ, KHOO C, DAIN JA, ROWLEY DC & SEERAM NP. 2016. Effect of cranberry (Vaccinium macrocarpon) oligosaccharides on the formation of advanced glycationend-products. J Berry Res 6: 149-158. https://doi.org/10.3233/JBR-160126.
  • TREED RD ET AL. 2019. Chronic pain as a symptom or a disease. Pain 160: 19-27. https://doi.org/ 10.1097/j.pain.0000000000001384.
  • UCHIYAMA M & MIHARA M. 1978. Determination of malonaldehyde precursor in tissues by thiobarbituric acid test. Anal Biochem 86: 271-278. https://doi.org/10.1016/0003-2697(78)90342-1.
  • VALERIO DA ET AL. 2009. Quercetin reduces inflammatory pain: inhibition of oxidative stress and cytokine production. J Nat Prod 72: 1975-1979. https://doi.org/10.1021/np900259y.
  • WANG S ET AL. 2019. Developmental neurotoxicity of reserpine exposure in zebrafish larvae (Danio rerio). Comparative Biochemistry and Physiology Part C: Regul Toxicol Pharmacol 223: 115-123. https://doi.org/10.1016/j.cbpc.2019.05.008.
  • WANG-YANG W, WU Y, HUANG H, HE C, LI W, WANG H, CHEN H & YIN Y. 2014. Biochanin A attenuates LPS-induced pro-inflammatory responses and inhibits the activation of the MAPK pathway in BV2 microglial cells. Int. J Mol Med 35: 391-398. https://doi.org/10.3892/ijmm.2014.2020.
  • WHISTLER WA & ELEVITCH CR. 2006. Syzygium malaccense. In: Elevitch CR (Ed), Species Profiles for Pacific Island Agroforestry. Holualoa, Hawai: Permanent Agriculture Resources (PAR), p. 1-13.
  • WILLIAMSON EM, OKPAKO DT & EVANS FJ. 1996. Selection, Preparation and Pharmacological Evaluation of Plant Material, Chichester: J Wiley & Sons, 238 p. ISBN: 978-0-471-94217-7.
  • YANG YL, LIU M, CHENG X, LI W, ZHANG S, WANG Y & DU G. 2019. Myricitrin blocks activation of NF-κB and MAPK signaling pathways to protect nigrostriatum neuron in LPS-stimulated mice. J Neuroimmunol 337: 577049. https://doi.org/10.1016/j.jneuroim.2019.577049.
  • ZHANG C, BROWNE A, CHILD D, DIVITO JR, STEVENSON JA & TANZI RE. 2010. Loss of function of ATXN1 increases amyloid beta-protein levels by potentiating beta-secretase processing of beta-amyloid precursor protein. J Biol Chem 285: 8515-8526. https://doi.org/10.1074/jbc.M109.079079.

Publication Dates

  • Publication in this collection
    26 Nov 2021
  • Date of issue
    2021

History

  • Received
    30 Mar 2021
  • Accepted
    21 Sept 2021
Academia Brasileira de Ciências Rua Anfilófio de Carvalho, 29, 3º andar, 20030-060 Rio de Janeiro RJ Brasil, Tel: +55 21 3907-8100 - Rio de Janeiro - RJ - Brazil
E-mail: aabc@abc.org.br