Acessibilidade / Reportar erro

Anti-Leishmania amazonensis activity of the marine sponge Dercitus (Stoeba) latex (Porifera) from São Pedro and São Paulo Archipelago, Pernambuco, Brazil

Abstract

The search for new therapeutic strategies for leishmaniasis treatment is essential due to the side effects of available drugs and the increasing incidence of resistance to them. Marine sponges use chemical compounds as a defense mechanism, and several of them present interesting pharmacological properties. The aim of this study was to evaluate the in vitro activity of the aqueous extract of the marine sponge Dercitus (Stoeba) latex against Leishmania amazonensis. MIC and toxicity against mammal cells were evaluated through broth microdilution assays. Transmission electron microscopy analysis was performed to assess possible effects on L. amazonensis ultrastructure. Arginase and proteolytic activities were measured by spectrometric methodologies. The extract of Dercitus (Stoeba) latex displayed antileishmanial activity and moderate toxicity against peritonial macrophages. Ultrastructural changes were observed after the growth of L. amazonensis promastigotes in the presence of the extract at 150 µg.ml-1 (IC50), mainly on acidocalcysomes. The extract was able to inhibit the activity of arginase and serine proteases. This study shows that Dercitus (Stoeba) latex aqueous extract may be a novel potential source of protozoa protease inhibitors and drugs that are less toxic to be used in the treatment of L. amazonensis infections.

Key words
Arginase; Dercitus (Stoeba) latex; Leishmania amazonensis ; leishmaniasis; proteolytic activity

INTRODUCTION

Leishmaniasis refers to a group of diseases caused by protozoa Leishmania and is transmitted by the bite of an infected sandfly. Clinically, the infectioncan progress in several ways, such as a chronic skin ulcer (cutaneous leishmaniasis), an erosive mucosal disease with severe facial disfigurement (mucocutaneous leishmaniasis), or a life-threatening systemic infection with hepato-splenomegaly and bone marrow involvement (visceral leishmaniasis) (Psicopo & Mallia 2009PSICOPO TV & MALLIA AC. 2009. Leishmaniasis. Postgrad Med J 83: 649-657., David & Craft 2009DAVID CV & CRAFT N. 2009. Cutaneous and mucocutaneous leishmaniasis. Dermatol Ther 22: 491-502.).

Leishmania species express molecules, such as proteases, that play different roles in host tissue invasion and immune evasion mechanisms (Soares et al. 2003SOARES RMA, SANTOS AL, BONALDO MC, ANDRADE AF, ALVIANO CS, ANGLUSTER J & GOLDENBERG S. 2003. Leishmania (Leishmania) amazonensis: differential expression of proteinases and cell-surface polypeptides in avirulent and virulent promastigotes. Exp Parasitol 104: 104-112., McKerrow 2018MCKERROW JH. 2018. The diverse roles of cysteine proteases in parasites and their suitability as drug targets. PLoS Negl Dis 12: e0005639.). Among them, arginase, a metalloenzyme involved in the hydrolysis of L-arginine and the regulation of nitric oxide synthesis, plays an important role in the subversion of macrophage function during the disease (Balaña-Fouce et al. 2012BALAÑA-FOUCE R, CALVO-ÁLVAREZ E, ÁLVAREZ-VELILLA R, PRADA CF, PÉREZ-PERTEJO Y & REGUERA RM. 2012. Role of trypanosomatid´s arginase in polyamine biosynthesis and pathogenesis. Mol Biochem Parasitol 181: 85-93.).

Pentavalent antimonials are the first-choice therapeutic regimen for the treatment of leishmaniasis. Whenever it does not work or cannot be prescribed, amphotericin B, pentamidine, or paromomycin serve as a second-choice line of treatment. Miltefosine, the only anti-Leishmania drug available for oral treatment, is normally prescribed for visceral leishmaniasis, and azoles, such as fluconazole, have been used for the treatment of the cutaneous form. However, the development of resistant strains has often led to therapeutic failure (Mans et al. 2016MANS DRA ET AL. 2016. Monitoring the response of patients with cutaneous leishmaniasis to treatment with pentamidine isethionate by quantitative real-time PCR, and identification of Leishmania parasites not responding to therapy. Clin Exp Dermatol 41: 610-615., Ponte-Sucre 2017PONTE SUCRE A, GAMARRO F, DUJARDIN JC, BARRETT MP, LÓPEZ-VÉLEZ R, GARCÍA-HERNÁNDEZ R, POUNTAIN AW, MWENECHANYA R & PAPADOPOULOU B. 2017. Drug resistance and treatment failure in leishmaniasis: a 21st century challenge. PLoS Negl Trop Dis 11: e0006052., Rodrigues et al. 2006RODRIGUES AM, HUEB M, SANTOS TARR & FONTES CJF. 2006. Factors associated with treatment failure of cutaneous leishmaniasis with meglumine antimoniate. Rev Soc Bras Med Trop 39: 139-145.).

The relevance of non-conventional drugs in the treatment of infectious diseases has increased remarkably in recent years. Marine organismsoffer an unprecedented opportunity for pharmacological exploitation, mainly because their metabolites have already been reported as rich sources of novel chemical compounds which may lead to more effective drugs (Haefner 2003HAEFNER B. 2003. Drugs from the deep: marine natural products as drug candidates. Drug Discov Today 8: 536-544., Mollica et al. 2012MOLLICA A, LOCATELLI M, STEFANUCCI A & PINNEN F. 2012. Synthesis and bioactivity of secondary metabolites from marine sponges containing dibrominated indolic systems. Molecules 17: 6083-6099.).

Sponges are marine, sessile, filter-feeding multicellular animals (without true organs) that use chemical compounds as a defense mechanism. Their extracts and compounds have been reported to present antibacterial (Mangalindan et al. 2000MANGALINDAN GC, TALAUE MT, CRUZ LJ, FRANZBLAU SG, ADAMS LB, RICHARDSON AD, IRELAND CM & CONCEPCION GP. 2000. Agelasine F from a Philippine Agelas sp. sponge exhibits in vitro antituberculosis activity. Planta Medical 66: 364-365.), antiviral (Da Silva et al. 2006DA SILVA AC ET AL. 2006. In vitro antiviral activity of marine sponges collected off Brazilian coast. Biol Pharm Bull 29: 135-140.), antifungal (Wattanadilok et al. 2007WATTANADILOK R, SAWANGWONG P, RODRIGUES C, CIDADE H, PINTO M, PINTO E, SILVA A & KIJJOA A. 2007. Antifungal activity evaluation of the constituents of Haliclona baeri and Haliclona cymaeformis, collected from the gulf of Thailand. Mar Drugs 5: 40-51.), and even antileishmanial activities, such as seen on those obtained from Cliona varians (Moura et al. 2006MOURA RM, QUEIROZ AFS, FOOK JMSLL, DIAS ASF, MONTEIRO NKV, RIBEIRO JKC, MOURA GEDD, MACEDO LLP, SANTOS EA & SALES MP. 2006. CvL, a lectin from marine sponge Cliona varians: Isolation, characterization and its effects on pathogenic bacteria and Leishmania promastigotes. Comp Biochem Physiol A Mol Integr Physiol 145: 517-523.), Haliclona exigua (Dube et al. 2007DUBE A, SINGH N, SAXENA A & LAKSHMI V. 2007. Antileishmanial potential of a marine sponge Haliclona exígua (Kirkpatrick) against experimental visceral leishmaniasis. Parasitol Res 101: 317-324.), Plakortis angulospiculatus (Kossuga et al. 2008KOSSUGA MH ET AL. 2008. Antiparasitic, antineuroinflamatory, and cytotoxic polypeptides from the marine sponge Plakortis angulospiculatus collected in Brazil. J Nat Prod 71: 334-339.), and Dragmaxia undata (Carballeira et al. 2011CARBALLEIRA NM, MONTANO N, CINTRÓN GA, MÁRQUEZ C, RUBIO CF, PRADA CF & BALAÑA-FOUCE R. 2011. First total synthesis and antileishmanial activity of (Z)-16-methyl-11-heptadecenoic acid, a new marine fatty acid from the sponge Dragmaxia undata. Chem Phys Lipids 164: 113-117.).

In the present study, the antileishmanial activity of the endemic brazilian marine sponge Dercitus (Stoeba) latex (Moraes & Muricy 2007MORAES F & MURICY MA. 2007. New species of Stoeba (Demospongiae: Astrophorida) from oceanic islands off north - eastern Brazil. J Mar Biol Ass UK 87: 1387-1393.) collected from São Pedro and São Paulo Archipelago was evaluated.

MATERIALS AND METHODS

Sponge sampling and identification

São Pedro and São Paulo Archipelago is one of the smallest and most isolated set of islands in the world, located 1000 km from the city of Natal, Rio Grande do Norte State, Northeastern Brazil. The sponge was manually collected by scuba diving (0˚55’N 29˚21’W) and preserved in 70% ethanol. The specimen was photographed in situ using a Nikonos V camera with 35 mm Nikkor lens and close-up kit. The positive film was digitalized using a Nikon Coolscan IV ED scanner (Moraes & Muricy 2007MORAES F & MURICY MA. 2007. New species of Stoeba (Demospongiae: Astrophorida) from oceanic islands off north - eastern Brazil. J Mar Biol Ass UK 87: 1387-1393.). Samples of the sponge were stored at -20˚ C for later evaluation.

Preparation of extracts

The sponge was washed with distilled water and transported to the laboratory under refrigeration. Then, it was kept in milli-Q water for 72 hours at 4o C for the extraction. Thus, the extract was lyophilized in Speed ​​Vac® and resuspended in sterile distilled water to a stock solution of 100 mg.ml-1.

Parasite

Leishmania (Leishmania) amazonensis Josefa strain (MHOM/BR/75/Josefa), originally isolated from a human case of cutaneous leishmaniasis, was kindly gifted by Dra Elvira Saraiva (Instituto de Microbiologia Paulo de Góes, UFRJ, Brazil). Promastigote forms were maintained by weekly transfers in 25-cm2 culture flasks with Schneider´s insect medium (Sigma Aldrich®, St. Louis, USA), pH 7.2, supplemented with 10% fetal calf serum (FCS) (Cultlab®, São Paulo, Brazil) and gentamicine sulphate (Schering-Plough®, São Paulo, Brazil) (80 µg.ml-1) at 26° C.

Cytotoxicity assay

Mice were subjected to intraperitoneal stimulation with thioglycolate for 96 hours. Then, peritoneal macrophages were obtained and incubated (5x105/well) on 96-well plates with various concentrations (promastigotes IC10, IC50 and MIC) of D. (S.) latex extract at 37o C in 5% CO2 for 24 h. In vitro cytotoxicity of the extract on peritoneal murine macrophages performed after intraperitoneal stimulation for 96 hours by thioglycolate was assessed by colorimetric 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reduction assay (Mosmann 1983MOSMANN J. 1983. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods 65: 55-63.). Firstly, macrophages were treated with 5 mg.ml-1 MTT and incubated for 3h at 37° C. Afterwards, dimethyl sulfoxide (DMSO) was added for 1 h to solubilize formazan crystals, and absorbance was measured at 540 nm. Cell viability was expressed as a percentage of average viable control cells. Three independent experiments were carried out in triplicates.Results were expressed as the concentration able to reduce cell viability by 50% (CC50), in comparison to positive control. This protocol was approved by ethical committee of the UFRJ Health Sciences Center under the number IMPG020.

Antileishmanial activity

Activity against L. amazonensis promastigotes

Cells (1x106 parasites.ml-1) were incubated at 26° C for 120 h in fresh medium (Schneider´s insect medium) supplemented with 10% FCS in the absence (control parasites) or presence (treated parasites) of several concentrations (2 µg.ml-1 to 1000 μg.ml-1) of D. (S.) latex aqueous extract. Glucantime (Sanofi-Aventis®) at 30 µg.ml-1 to 470 µg.ml-1 and Amphotericin B at 3 µg.ml-1 to 50 µg.ml-1 were used as control drugs. Parasite viability was assessed before and after incubation by evaluating cell motility and trypan blue staining. The 100% inhibitory concentration (MIC) and 50% inhibitory concentration (IC50) were determined by linear regression analysis using Microsoft Excel® (Microsoft Corporation, WA, USA).

Activity against L. amazonensis amastigotes

Peritoneal macrophages adhered to glass coverslips were washed with PBS pH 7.2 and incubated with promastigotes (5x106 parasites.ml-1) at stationary growth phase in DMEM medium (Thermo Fisher Scientific, MA, USA) supplemented with 2% FCS at 37° C in 5% CO2 for one hour. After interaction, cells were washed in PBS and incubated with DMEM supplemented with 10% FCS in the presence or absence of D. (S.) latex extract at promastigotes IC50. After 24h and 48h, the material was washed, fixed with methanol for 5 minutes, dehydrated with acetone-xylol, and stained with 36% Giemsa. Infected macrophages were visualized by light microscopy and counted. Three independent experiments were carried out in triplicate.

Transmission electron microscopy

Parasites incubated for 120 h in the presence or absence of D. (S.) latex aqueous extract at promastigotes IC50 were fixed in 4% formaldehyde, 2.5 % glutaraldehyde, and 0.1 M sodium cacodylate buffer (pH 7.2) for 1h at room temperature. Cells were post-fixed in 1% OsO4, dehydrated and embedded as previously described (Bisaggio et al. 2006BISAGGIO DF, CAMPANATI L, PINTO RC & SOUTO-PADRÓN T. 2006. Effect of suramin on trypomastigote forms of Trypanossoma cruzi: changes on cell motility and on the ultra structure of the flagellum-cell body attachment region. Acta Tropica 98: 162-175.). Ultrathin sections obtained with a Reichert UltraCut S ultra microtome were stained with uranyl acetate and then analyzed in a FEI Morgagni F 268 transmission electron microscope, operating at 80 kV, equipped with a Megaview G2 Camera. Alternatively, cells were grown in the presence or absence of IC50 extract for 72h, washed in PBS and placed on Formvar®-coated grids (Agar Scientific, United Kingdom) and subsequently observed in the transmission electron microscope. A descriptive analysis of morphometric parameters was performed using ImageJ® (National Institutes of Health, MD, USA). Morphometric parameters such as number, diameter, circularity and absolute volume were measured.

Determination of arginase activity

Arginase enzyme activity was measured as previously described (Kropf et al. 2005KROPF P, FUENTES JM, FÄHNRICH E, ARPA L, HERATH S, WEBER V, SOLER G, CELADA A, MODOLELL M & MÜLLER I. 2005. Arginase and polyamine synthesis are key factors in the regulation of experimental leishmaniasis in vivo. The FASEB J 19: 1000-1002.), with slight modifications. Promastigotes were incubated for 72 h in the absence or presence of D. (S.) latex aqueous extract at promastigotes IC50 concentration. The parasites were lysed with 0.1% Triton X-100, and then 25 mM Tris-HCl pH 8.3 was added. After this, 10 mM MnCl2 was added, and the enzyme was activated by heating for 10 min at 56° C. Arginine hydrolysis was conducted by incubating the lysate with 0.5 M L-arginine (pH 9.7) at 37° C for 15-20 min. The reaction was stopped with the addition of an acidic solution (H2SO4 (96%)/H3PO4 (85%)/H2O (1/3/7, v/v/v). Urea concentration was measured at 540 nm in spectrophotometer (FLUOstar OPTIMA, BMG Labtech, Offenburg, Germany) after addition of α-isonitrosopropiophenone (dissolved in 100% ethanol) followed by heating at 95° C for 30 min.

Anti-Proteolytic activity

The effect of D. (S.) latex on the proteolytic activity of L. amazonensis was assessed as previously described (Kamboj et al. 1993KAMBOJ RC, PAL S, RAGHAY N & SINGH H. 1993. A selective colorimetric assay for cathepsin L using Z-Phe-Arg-4-methoxy-beta-naphthylamide. Biochimie 75: 873-878.), using the fluorogenic substrate Z-Phe-Arg-4-methoxy-β-naphthylamide (Sigma Aldrich®, St. Louis, USA). Briefly, 40 µg of a proteic extract obtained from L. amazonensis and 20 µg of trypsin were diluted in 50mM phosphate buffer pH 5.5. Then, 2.5 µl of the fluorogenic substrate Z-Phe-Arg-4-methoxy-naphthylamide was added and incubated at 37o C for 1 hour with [1-[N-[(L-3-trans-carboxyoxirane-2-carbonyl)-L-leucyl]amino]-4-guanidinobutane] (E64), phenylmethylsulfonyl fluoride (PMSF), or D. (S.) latex aqueous extract. The reaction was stopped by the addition of 150 µl of 5% trichloroacetic acid. Substrate cleavage was measured at 380 and 450 nm for fluorescence excitation and emission, respectively.

Statistical analysis

All experiments were repeated at least three times, all systems were performed in triplicate sets, and the results were expressed as means ± standard deviation. Data were analysed by Student´s t-test, and P<0.05 was considered statistically significant.

RESULTS

Antileishmanial activity and cytotoxicity assay

D. (S.) latex aqueous extract inhibited the growth of L. amazonensis promastigotes in a dose dependent manner (data not shown), reaching absolute inhibition at 372 μg.ml-1 and the 50% inhibitory concentration (IC50) was 150 µg.ml-1. The control drugs Amphotericin B and Glucantime exhibited IC50 of 4 µg.ml-1 and 60 µg.ml-1, respectively. The IC50 of sponge extract against L. amazonensis promastigotes, its CC50 against mouse peritoneal macrophages, and the selective indexes are presented in Table I.

Table I
Inhibition of promastigote growth and peritoneal murine macrophages citotoxicity by D. (S.) latex aqueous extract.

Treatment with the IC50 of the sponge extract reduced the macrophage infection by 32.6% within 24 hours, and no significant difference was observed after a 48h-incubation (data not shown). Furthermore, it was observed a 21% reduction in the number of amastigotes per macrophage.

Transmission electron microscopy

Electron microscopy analysis (Figure 1) showed different degrees of damage in treated parasites, including the discontinuity of the nuclear membrane (Figure 1d, f, black arrows), formation of concentric structures with membranes (Figure 1e, black arrow) and the presence of several axonemes (at least three) in the cytoplasm (Figure 1g, black arrow). These findings indicate the presence of multiple flagella or the internalization of a single one, probably coiled-appearing several times in the cut, suggesting effects on the cell cycle. Changes in the electron density of acidocalcisomes were also observed (Figure 1h, i, black arrows). Moreover, descriptive analysis of morphometric parameters revealed an increase in the number of acidocalcisomes and the circularity of these organelles after a 72h-incubation with the IC50 extract (Figure 2 and Table II).

Figure 1
Analysis by TEM of L. amazonensis promastigotes in absence (a-c) or presence (d-i) of IC50 D. (S.) latex aqueous extract. Different degrees of damage were observed in treated cells, including the discontinuity of the nuclear membrane (d and f arrowhead), formation of concentric structures with membranes (e, black arrow), and the presence of several axonemes (at least three) in the cytoplasm (g, black arrow). Changes in the electron density of acidocalcisomes were also observed (h, i, black arrows).
Figure 2
Numeric analysis of the acidocalcisomes in L. amazonensis. The number of the organelles was analysed and compared between control and treated cells. The cells treated with the sponge aqueous extract were subdivided into two groups representing cells between 11 and 20, and more than 20 acidocalcisomes per cell. Results are expressed in percentage of cell populations.
Table II
Numerical and circular changes in acidocalcisomes of L. amazonensis promastigotes grown in the presence of the extract IC50 for 72 hours. The (*) represent statistically significant data.

Determination of arginase activity

Arginase activity was reduced in 28.66% in the presence of the sponge aqueous extract as demonstrated by the results shown in Figure 3.

Figure 3
Effect of the aqueous extract of Dercitus (Stoeba) latex on arginase activity of L. amazonensis promastigotes. Parasites were grown on Schneider medium at 26 oC for 72 h in the presence of 150 µg.ml-1 Dercitus (Stoeba) latex aqueous extract. Then, parasites were washed, lysed, and arginase activity was measured as described at Materials and Methods section. Values represent means +/- standard deviation of three different experiments. (*): P < 0.05.

Anti-Proteolytic activity

Data obtained revealed that the incubation of the protein cell extract with BSA in phosphate buffer pH 5.5 led to an inhibition of 35% of the proteolytic activity. This result was better than that observed with PMSF, a classic serine protease inhibitor (data not shown). Then, an assay was performed in order to analyze the hydrolysis of a specific fluorogenic substrate for serine protease. When the cysteine ​​proteases were inhibited by E64, it was observed the inhibition of fluorescence emission when PMSF or aqueous extract of D. (S.) latex (IC50) were added to the system (Figure 4). Treatment with E64 led to a significant hydrolysis of the fluorogenic substrate. Moreover, treatment with PMSF and aqueous extract D. (S.) latex significantly decreased substrate cleavage.

Figure 4
Effect of Dercitus (Stoeba) latex aqueous extract on the proteolytic activity of L. amazonensis. Hydrolysis of the fluorogenic substrate Z-Phe-Arg 4-methoxy-β-naphthylamide by L. amazonensis (LA) promastigotes cell extract in the presence or absence of proteolytic inhibitors PMSF and/or E64 and/or D. (S.) latex aqueous extract. Trypsin was used as control. The results represent the average of three experiments in triplicate ± standard deviation. (*): P < 0.05.

DISCUSSION

Natural products have been successfully used in the search for compounds with anti-Leishmania activity (da Silva et al. 2018DA SILVA BJM, HAGE AAP, SILVA EO & RODRIGUES APD. 2018. Medicinal plants from the Brazilian Amazonian region and their antileishmanial activity: a review. J Integr Med 16: 211-222.). Although most of natural compounds with antimicrobial activity are derived from plant, the use of marine substances seems to be very promising (Donia & Hamman 2003DONIA M & HAMMAN MT. 2003. Marine natural products and their potential applications as anti-infective agents. Lancet Infect Dis 3: 338-348., Laport et al. 2009LAPORT M, SANTOS OCS & MURICY G. 2009. Marine sponges: potential sources of new antimicrobial drugs. Curr Pharm Biotechnol 10: 86-105.). In this study, D. (S.) latex aqueous extract presented antileishmanial activity against promastigote forms, and microscopy analysis revealed damage on several structures, including acidocalcisomes. These structures are dense acidic organelles with a high concentration of phosphorus, presented mostly in the form of polyphosphates complexed with calcium and other elements. Several functions have been attributed to the acidocalcisomes, such as the storage of high energy compounds, calcium and other cations, and the regulation of the intracellular pH and osmolarity (DoCampo & Huang 2015DOCAMPO R & HUANG G. 2015. Calcium signaling in trypanosomatid parasites. Cell Calcium 57: 194-202.). Despite the changes in the number and shape of acidocalcisomes revealed in this study, protozoan susceptibility to antimicrobial drugs seems to be unrelated, as demonstrated by the use of sitamaquine (López-Martín et al. 2008LÓPEZ-MARTÍN C, PÉREZ-VICTORIA JM, CARVALHO L, CASTANYS S & GAMARRO F. 2008. Sitamaquine sensitivity in Leishmania species is not mediated by drug accumulation in acidocalcisomes. Antimicrob Agents Chemother 52: 4030-4036.). However, a recent study using nelfinavir, an HIV protease inhibitor, reported the accumulation of this drug in acidocalcisomes of resistant strains of L. amazonensis, suggesting that an increase in the number of this vesicle might be considered as a resistant profile feature (Kumar et al. 2013KUMAR P, LODGE R, RAYMOND F, RITT JF, JALAGUIER P, CORBEIL J, OUELLETTE M & TREMBLAY MJ. 2013. Gene expression modulation and the molecular mechanisms involved in Nelfinavir resistance in Leishmania donovani axenic amastigotes. Mol Microbiol 89: 565-582.). Furthermore, an inositol triphosphate receptor located in acidocalcisomes of Trypanosoma brucei was recently identified and considered essential for the growth and the establishment of an efficient animal infection (Huang et al. 2013HUANG G, BARTLETT PJ, THOMAS AP, MORENO SN & DOCAMPO R. 2013. Acidocalcisomes of Trypanosoma brucei have an inositol 1,4,5-trisphosphate receptor that is required for growth and infectivity. Proc Natl Acad Sci USA 110: 1887-1892.). All these characteristics point to acidocalcisome as an important multifunctional organelle associated to crucial biological processes among protozoa.

The ability to survive and multiply within macrophages is a feature of several infectious agents, including Trypanosoma cruzi and Leishmania spp. In order to sustain a chronic infection, parasites must subvert macrophage-accessory cell activities and ablate the development of protective immunity (Alexander et al. 2002ALEXANDER J, SATOSKAR AR &RUSSEL DG. 2002. Leishmania species: models of intracellular parasitism. J Cell Sci 112: 2993-3002.). Nevertheless, the most important mechanism for the killing of Leishmania and leishmaniasis control is the production of nitric oxide by macrophages (Holzmuller et al. 2006HOLZMULLER P, BRAZ-GONÇALVES R & LEMESRE JL. 2006. Phenotypical characteristics, biochemical pathways, molecular targets and putative role of nitric oxide-mediated programmed cell death in Leishmania. Parasitology 132: S19-S32.). The inhibition of arginase activity may influence parasite viability in L. major and L. infantum–infected macrophages (Iniesta et al. 2001INIESTA V, GÓMES-NIETO LC & CORRALIZA I. 2001. The inhibition of arginase by Nω- Hydroxy-L-Arginine controls the growth of Leishmania inside macrophages. J Exp Med 193: 777-783.). It is postulated that among Leishmania species, arginase activity must modulate nitric oxide synthase activity by using L-arginine, which is a common substrate for enzymatic activities (Boucher et al. 1999BOUCHER JL, MOALI C & TENU JP. 1999. Nitric oxide biosynthesis, nitric oxide synthase inhibitors and arginase competition for L-arginine utilization. Cell Mol Life Sci 55: 1015-1028., Wanasen et al. 2008WANASEN N & SOONG L. 2008. L-arginine metabolism and its impact on host immunity against Leishmania infection. Immunol Res 41: 15-25.). In L. amazonensis, the arginase protein is concentrated in glycosomes both in promastigotes and amastigotes, suggesting that its location is important for enzyme activity, which modulates the L-arginine intracellular levels. Knockout parasites were able to infect macrophages but were not able to sustain the infection. It could be explained by a failure in modulating the availability of L-arginine to host cell and leading to an increased production of NO by infected macrophages (Da Silva et al. 2008DA SILVA ER, DA SILVA MF, FISCHER H, MORTARA RA, MAYER MG, FRAMESQUI K, SILBER AM & FLOETER-WINTER LM. 2008. Biochemical and biophysical properties of a highly active recombinant arginase from Leishmania (Leishmania) amazonensis and subcellular localization of native enzyme. Mol Biochem Parasitol 159: 104-111.). Promisingly, besides the effect on acidocalcisomes, the sponge aqueous extract was able to reduce arginase activity of L. amazonensis promastigotes, reinforcing the ability of this extract of modulating distinct mechanisms of L. amazonensis virulence.

Analysis of proteolytic enzymes of pathogenic organisms might lead to the design of powerful chemotherapeutic agents against these microorganisms (Grandgenett et al. 2007GRANDGENETT PM, OTSU K, WILSON HR, WILSON ME & DONELSON JE. 2007. A function for a specific zinc Metalloprotease of African Trypanosomes. PLoS Pathog 3: 1432-1445., Casgrain et al. 2016CASGRAIN PA, MARTEL C, MCMASTER WR, MOTTRAM JC, OLIVIER M & DESCOTEAUX A. 2016. Cysteine peptidase B regulates Leishmania mexicana virulence through the modulation of GP63 expression. PLoS Pathog 12: e1005658.). The most studied classes of proteases in Leishmania spp. are cysteine-, metallo-, and serine proteases, since they are directly related to virulence. These enzymes cleave host proteins, neutralize immune response, disrupt fibronectin and extracellular matrix, overall enhancing promastigotes infectivity (Mottram et al. 2004MOTTRAM JC, COOMBS GH & ALEXANDER J. 2004. Cysteine peptidases as virulence factors of Leishmania. Curr Opin Microbiol 7: 375-381.). Then, proteases promote the survival of the parasite and the interaction with host tissues (Silva-Lopez et al. 2005SILVA-LOPEZ RE, PINTO-COELHO MG & DE SIMONE G. 2005. Characterization of an extracellular serine protease of Leishmania (Leishmania) amazonensis. Parasitology 131: 85-96.). Results obtained suggest that D. (S.) latex aqueous extract may inhibit L. amazonensis serine proteases activity. Silva-Lopez and colleagues (2007) showed that the serine proteases inhibitors TPCK, benzamidine, and (ShPI-1), isolated from the sea anemone Stichodactyla heliantus affected cell viability and caused ultrastructural changes in the flagellar pocket, changes in membrane parasites, and the formation of intracellular vesicular bodies in L. amazonensis (Silva-Lopez et al. 2007SILVA-LOPEZ RE, MORGADO-DÍAZ JÁ, CHÁVEZ MA & GIOVANNI-DE-SIMONE S. 2007. Effects of serine protease inhibitors on viability and morphology of Leishmania (Leishmania) amazonensis promastigotes. Parasitol Res 101: 1627-1635.). Nogueira and coworkers (2013) also showed a potent serine protease inhibitor purified from sea anemone which showed significant anti–T. cruzi and moderate anti–L. amazonensis activities (Nogueira et al. 2013NOGUEIRA RC, ROCHA VP, NONATO FR, TOMASSINI TC, RIBEIRO IM, DOS SANTOS RR & SOARES MB. 2013. Genotoxicity and antileishmanial activity evaluation of Physalis angulata concentrated ethanolic extract. Environ Toxicol Pharmacol 36: 1304-1311.).

From the results obtained in our study, it was observed that the aqueous extract of D. (S.) latex has antileishmanial activity, since it was able to disrupt meaningful cellular components and inhibit enzymes associated with virulence in the parasite. The study of the extract of D. (S.) latex is promising and contributes to the advancement in the search for new, more effective, and less toxic drugs that can be used in the treatment of cutaneous leishmaniasis. Furthermore, this study shows that marine animals may be a source of protozoa protease inhibitors. The limitation of a study carried out with extracts in which there is no isolation and identification of compounds is understood; however the contribution to the confirmation of the potential of marine sponges as a source of new drugs is considered.

ACKNOWLEDGMENTS

We would like to thank the Marinha do Brasil and Secretaria da Comissão Interministerial para Recursos do Mar (SECIRM) for providing logistic support for sponge collection. This research was supported by the Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) and the Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ). This study was also financed in part by the Coordenação de Aperfeiçoamento de Pessoal de Nível Superior – Brazil (CAPES) – Finance Code 001.

REFERENCES

  • ALEXANDER J, SATOSKAR AR &RUSSEL DG. 2002. Leishmania species: models of intracellular parasitism. J Cell Sci 112: 2993-3002.
  • BALAÑA-FOUCE R, CALVO-ÁLVAREZ E, ÁLVAREZ-VELILLA R, PRADA CF, PÉREZ-PERTEJO Y & REGUERA RM. 2012. Role of trypanosomatid´s arginase in polyamine biosynthesis and pathogenesis. Mol Biochem Parasitol 181: 85-93.
  • BISAGGIO DF, CAMPANATI L, PINTO RC & SOUTO-PADRÓN T. 2006. Effect of suramin on trypomastigote forms of Trypanossoma cruzi: changes on cell motility and on the ultra structure of the flagellum-cell body attachment region. Acta Tropica 98: 162-175.
  • BOUCHER JL, MOALI C & TENU JP. 1999. Nitric oxide biosynthesis, nitric oxide synthase inhibitors and arginase competition for L-arginine utilization. Cell Mol Life Sci 55: 1015-1028.
  • CARBALLEIRA NM, MONTANO N, CINTRÓN GA, MÁRQUEZ C, RUBIO CF, PRADA CF & BALAÑA-FOUCE R. 2011. First total synthesis and antileishmanial activity of (Z)-16-methyl-11-heptadecenoic acid, a new marine fatty acid from the sponge Dragmaxia undata. Chem Phys Lipids 164: 113-117.
  • CASGRAIN PA, MARTEL C, MCMASTER WR, MOTTRAM JC, OLIVIER M & DESCOTEAUX A. 2016. Cysteine peptidase B regulates Leishmania mexicana virulence through the modulation of GP63 expression. PLoS Pathog 12: e1005658.
  • DA SILVA AC ET AL. 2006. In vitro antiviral activity of marine sponges collected off Brazilian coast. Biol Pharm Bull 29: 135-140.
  • DA SILVA BJM, HAGE AAP, SILVA EO & RODRIGUES APD. 2018. Medicinal plants from the Brazilian Amazonian region and their antileishmanial activity: a review. J Integr Med 16: 211-222.
  • DA SILVA ER, DA SILVA MF, FISCHER H, MORTARA RA, MAYER MG, FRAMESQUI K, SILBER AM & FLOETER-WINTER LM. 2008. Biochemical and biophysical properties of a highly active recombinant arginase from Leishmania (Leishmania) amazonensis and subcellular localization of native enzyme. Mol Biochem Parasitol 159: 104-111.
  • DAVID CV & CRAFT N. 2009. Cutaneous and mucocutaneous leishmaniasis. Dermatol Ther 22: 491-502.
  • DOCAMPO R & HUANG G. 2015. Calcium signaling in trypanosomatid parasites. Cell Calcium 57: 194-202.
  • DONIA M & HAMMAN MT. 2003. Marine natural products and their potential applications as anti-infective agents. Lancet Infect Dis 3: 338-348.
  • DUBE A, SINGH N, SAXENA A & LAKSHMI V. 2007. Antileishmanial potential of a marine sponge Haliclona exígua (Kirkpatrick) against experimental visceral leishmaniasis. Parasitol Res 101: 317-324.
  • GRANDGENETT PM, OTSU K, WILSON HR, WILSON ME & DONELSON JE. 2007. A function for a specific zinc Metalloprotease of African Trypanosomes. PLoS Pathog 3: 1432-1445.
  • HAEFNER B. 2003. Drugs from the deep: marine natural products as drug candidates. Drug Discov Today 8: 536-544.
  • HOLZMULLER P, BRAZ-GONÇALVES R & LEMESRE JL. 2006. Phenotypical characteristics, biochemical pathways, molecular targets and putative role of nitric oxide-mediated programmed cell death in Leishmania. Parasitology 132: S19-S32.
  • HUANG G, BARTLETT PJ, THOMAS AP, MORENO SN & DOCAMPO R. 2013. Acidocalcisomes of Trypanosoma brucei have an inositol 1,4,5-trisphosphate receptor that is required for growth and infectivity. Proc Natl Acad Sci USA 110: 1887-1892.
  • INIESTA V, GÓMES-NIETO LC & CORRALIZA I. 2001. The inhibition of arginase by Nω- Hydroxy-L-Arginine controls the growth of Leishmania inside macrophages. J Exp Med 193: 777-783.
  • KAMBOJ RC, PAL S, RAGHAY N & SINGH H. 1993. A selective colorimetric assay for cathepsin L using Z-Phe-Arg-4-methoxy-beta-naphthylamide. Biochimie 75: 873-878.
  • KOSSUGA MH ET AL. 2008. Antiparasitic, antineuroinflamatory, and cytotoxic polypeptides from the marine sponge Plakortis angulospiculatus collected in Brazil. J Nat Prod 71: 334-339.
  • KROPF P, FUENTES JM, FÄHNRICH E, ARPA L, HERATH S, WEBER V, SOLER G, CELADA A, MODOLELL M & MÜLLER I. 2005. Arginase and polyamine synthesis are key factors in the regulation of experimental leishmaniasis in vivo. The FASEB J 19: 1000-1002.
  • KUMAR P, LODGE R, RAYMOND F, RITT JF, JALAGUIER P, CORBEIL J, OUELLETTE M & TREMBLAY MJ. 2013. Gene expression modulation and the molecular mechanisms involved in Nelfinavir resistance in Leishmania donovani axenic amastigotes. Mol Microbiol 89: 565-582.
  • LAPORT M, SANTOS OCS & MURICY G. 2009. Marine sponges: potential sources of new antimicrobial drugs. Curr Pharm Biotechnol 10: 86-105.
  • LÓPEZ-MARTÍN C, PÉREZ-VICTORIA JM, CARVALHO L, CASTANYS S & GAMARRO F. 2008. Sitamaquine sensitivity in Leishmania species is not mediated by drug accumulation in acidocalcisomes. Antimicrob Agents Chemother 52: 4030-4036.
  • MANGALINDAN GC, TALAUE MT, CRUZ LJ, FRANZBLAU SG, ADAMS LB, RICHARDSON AD, IRELAND CM & CONCEPCION GP. 2000. Agelasine F from a Philippine Agelas sp. sponge exhibits in vitro antituberculosis activity. Planta Medical 66: 364-365.
  • MANS DRA ET AL. 2016. Monitoring the response of patients with cutaneous leishmaniasis to treatment with pentamidine isethionate by quantitative real-time PCR, and identification of Leishmania parasites not responding to therapy. Clin Exp Dermatol 41: 610-615.
  • MCKERROW JH. 2018. The diverse roles of cysteine proteases in parasites and their suitability as drug targets. PLoS Negl Dis 12: e0005639.
  • MOLLICA A, LOCATELLI M, STEFANUCCI A & PINNEN F. 2012. Synthesis and bioactivity of secondary metabolites from marine sponges containing dibrominated indolic systems. Molecules 17: 6083-6099.
  • MORAES F & MURICY MA. 2007. New species of Stoeba (Demospongiae: Astrophorida) from oceanic islands off north - eastern Brazil. J Mar Biol Ass UK 87: 1387-1393.
  • MOSMANN J. 1983. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods 65: 55-63.
  • MOTTRAM JC, COOMBS GH & ALEXANDER J. 2004. Cysteine peptidases as virulence factors of Leishmania. Curr Opin Microbiol 7: 375-381.
  • MOURA RM, QUEIROZ AFS, FOOK JMSLL, DIAS ASF, MONTEIRO NKV, RIBEIRO JKC, MOURA GEDD, MACEDO LLP, SANTOS EA & SALES MP. 2006. CvL, a lectin from marine sponge Cliona varians: Isolation, characterization and its effects on pathogenic bacteria and Leishmania promastigotes. Comp Biochem Physiol A Mol Integr Physiol 145: 517-523.
  • NOGUEIRA RC, ROCHA VP, NONATO FR, TOMASSINI TC, RIBEIRO IM, DOS SANTOS RR & SOARES MB. 2013. Genotoxicity and antileishmanial activity evaluation of Physalis angulata concentrated ethanolic extract. Environ Toxicol Pharmacol 36: 1304-1311.
  • PONTE SUCRE A, GAMARRO F, DUJARDIN JC, BARRETT MP, LÓPEZ-VÉLEZ R, GARCÍA-HERNÁNDEZ R, POUNTAIN AW, MWENECHANYA R & PAPADOPOULOU B. 2017. Drug resistance and treatment failure in leishmaniasis: a 21st century challenge. PLoS Negl Trop Dis 11: e0006052.
  • PSICOPO TV & MALLIA AC. 2009. Leishmaniasis. Postgrad Med J 83: 649-657.
  • RODRIGUES AM, HUEB M, SANTOS TARR & FONTES CJF. 2006. Factors associated with treatment failure of cutaneous leishmaniasis with meglumine antimoniate. Rev Soc Bras Med Trop 39: 139-145.
  • SILVA-LOPEZ RE, MORGADO-DÍAZ JÁ, CHÁVEZ MA & GIOVANNI-DE-SIMONE S. 2007. Effects of serine protease inhibitors on viability and morphology of Leishmania (Leishmania) amazonensis promastigotes. Parasitol Res 101: 1627-1635.
  • SILVA-LOPEZ RE, PINTO-COELHO MG & DE SIMONE G. 2005. Characterization of an extracellular serine protease of Leishmania (Leishmania) amazonensis. Parasitology 131: 85-96.
  • SOARES RMA, SANTOS AL, BONALDO MC, ANDRADE AF, ALVIANO CS, ANGLUSTER J & GOLDENBERG S. 2003. Leishmania (Leishmania) amazonensis: differential expression of proteinases and cell-surface polypeptides in avirulent and virulent promastigotes. Exp Parasitol 104: 104-112.
  • WANASEN N & SOONG L. 2008. L-arginine metabolism and its impact on host immunity against Leishmania infection. Immunol Res 41: 15-25.
  • WATTANADILOK R, SAWANGWONG P, RODRIGUES C, CIDADE H, PINTO M, PINTO E, SILVA A & KIJJOA A. 2007. Antifungal activity evaluation of the constituents of Haliclona baeri and Haliclona cymaeformis, collected from the gulf of Thailand. Mar Drugs 5: 40-51.

Publication Dates

  • Publication in this collection
    17 Oct 2022
  • Date of issue
    2022

History

  • Received
    03 Aug 2021
  • Accepted
    19 Nov 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