Accepted Manuscript Cytotoxicity of the coagulant Moringa oleifera lectin (cMoL) to B16-F10 melanoma cells
Luciana de Andrade Luz, Franco Aparecido Rossato, Rute Alves Pereira e Costa, Thiago Henrique Napoleão, Patrícia Maria Guedes Paiva, Luana Cassandra Breitenbach Barroso Coelho PII: DOI: Reference:
S0887-2333(17)30168-6 doi: 10.1016/j.tiv.2017.06.019 TIV 4038
To appear in:
Toxicology in Vitro
Received date: Revised date: Accepted date:
24 February 2017 16 June 2017 19 June 2017
Please cite this article as: Luciana de Andrade Luz, Franco Aparecido Rossato, Rute Alves Pereira e Costa, Thiago Henrique Napoleão, Patrícia Maria Guedes Paiva, Luana Cassandra Breitenbach Barroso Coelho , Cytotoxicity of the coagulant Moringa oleifera lectin (cMoL) to B16-F10 melanoma cells, Toxicology in Vitro (2017), doi: 10.1016/ j.tiv.2017.06.019
This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT Cytotoxicity of the coagulant Moringa oleifera lectin (cMoL) to B16-F10 melanoma cells
Luciana de Andrade Luza,*, Franco Aparecido Rossatob, Rute Alves Pereira e Costab, Thiago Henrique Napoleãoa, Patrícia Maria Guedes Paivaa, Luana Cassandra Breitenbach Barroso
Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco,
RI
a
PT
Coelhoa,*
b
SC
50670-901 Recife, Pernambuco, Brazil.
Departamento de Patologia Clínica, Faculdade de Ciências Médicas, Universidade Estadual
NU
de Campinas, 13083-887 Campinas, São Paulo, Brazil.
E-mail
addresses:
MA
*Corresponding author. Tel: +558121268540; fax: +558121268576.
[email protected]
Luz),
[email protected]
AC
CE
PT E
D
(L.C.B.B. Coelho).
(L.A.
1
ACCEPTED MANUSCRIPT ABSTRACT
Moringa oleifera seeds are used in alternative medicine to treat inflammation, tumors and bacterial and protozoan infections, for example. The seeds contain lectins, which are carbohydrate-binding proteins with several biological properties including cytotoxicity to
PT
cancer cells. In this work, we examined the cytotoxicity of the coagulant M. oleifera lectin
RI
(cMoL) on B16-F10 murine melanoma cells. cMoL cytotoxic effects were evaluated through
SC
trypan blue assay and flow cytometry analysis. Mitochondrial superoxide levels and activation of caspases 3, 8 and 9 were measured. cMoL (1.5–16 µM) reduced viability and
NU
caused cell death of B16-F10 cells with an IC50 of 9.72 µM. Flow cytometry analysis indicated induction of necrosis and suggested the presence of cells in late apoptosis.
MA
Specificity for tumor cells was observed since death of normal human fibroblasts (GN) was not higher than 20% in treatments with cMoL from 1.5 to 16 µM. Microscopy images
D
revealed rounded shape and reduction of volume in B16-F10 cells treated with cMoL. cMoL
PT E
increased mitochondrial ROS production and promoted caspases 3, 8 and 9 activation in B16F10 cells, indicating the activation of apoptosis-related pathway. In conclusion, this study
CE
demonstrates that cMoL is cytotoxic to B16-F10 cells, which stimulates more investigation on
AC
the anticancer potential of this lectin.
Keywords: Moringa oleifera; lectin; cytotoxicity; melanoma.
2
ACCEPTED MANUSCRIPT 1. Introduction
Moringa oleifera Lam. (Moringaceae) is a medium-sized tree autochthonous from northeastern India and widely distributed worldwide throughout the tropics and subtropics [1]. In developing countries, it is a vegetable and medicinal plant, and is source of oil with uses in
PT
industry and culinary [2]. It has been reported the pharmacological potential of M. oleifera as
RI
source of anxiolytic, antiepileptic, anticancer, antiulcer, anti-obesity and anti-inflammatory
SC
agents [3–9].
M. oleifera seeds are broadly used in water treatment for human consumption due to
NU
their coagulant properties [2]. Two lectins deemed cMoL (coagulant M. oleifera lectin) and WSMoL (water-soluble M. oleifera lectin) are among the coagulant proteins present in the
MA
seeds [10,11]. Lectins are proteins of non-immune origin that interact reversibly and specifically with sugars; this carbohydrate-binding effect endows diverse biological properties
D
to lectins, including the ability to alter the functioning of cells [12]. The cMoL is a heat-stable
PT E
and basic lectin (theoretical pI of 11.67) with a native molecular mass of 30 kDa, composed by subunits with 101 amino acids, and belonging to the α/β tertiary structure class. It is able to
CE
recognize both monosaccharides and oligosaccharide moieties of glycoproteins, with highest affinity for glucose, galactose, asialofetuin and azocasein [10,13]. This lectin demonstrated
AC
anticoagulant properties on hemostatic parameters of human blood coagulation [13] as well as insecticidal activity against Anagasta kuehniella [14]. However, no study on the anticancer properties of cMoL was performed until now. Mitochondria play an important role in cell death process since they regulate energy production and execution of cell death [15]. Mitochondria are also important in generating reactive oxygen and nitrogen species and are involved in pathways linked to cell proliferation, intracellular death signaling and disease pathogenesis [16]. Lectins have been able to interfere 3
ACCEPTED MANUSCRIPT with mitochondria function causing cell death. The Cratylia mollis seed lectin induced death of Trypanosoma cruzi epimastigote by promoting mitochondrial Ca2+ overload and stimulation of reactive oxygen species (ROS) production followed by necrosis-like cell death [17]. The Canavalia virosa seed lectin was toxic to rat C6 glioma cells by disrupting the mitochondrial membrane potential [18]. The treatment of HT29 human colon adenocarcinoma
PT
cells with a lectin from Bothrops jararacussu venom lead to mitochondrial respiration
RI
decrease and increase of cytochrome c release [19].
SC
In the present work, we evaluated the in vitro cytotoxic effects of cMoL to B16-F10 murine melanoma tumor cells and to normal human fibroblasts (GN) as well as mechanisms
NU
involved in B16-F10 cell death. In the context of studies searching for positive effects against cancer cells, this cell line was selected since it is very aggressive and presents high capacity
MA
for metastatic dissemination [20]. Some of the experiments were also conducted with normal fibroblasts obtained from gingival biopsy, since they correspond to untransformed primary
D
cells, which brings them very close to a physiological situation. In addition, these cells were
PT E
chosen due to their different origin being appropriate to test the specificity of the lectin
CE
cytotoxicity.
AC
2. Materials and methods
2.1. Plant material
The seeds of M. oleifera were collected in Recife City, State of Pernambuco, northeastern Brazil and stored at -20°C. A sample of the collected material is archived as voucher specimen (number 73,345) at the herbarium Dárdano de Andrade Lima from the Instituto Agronômico de Pernambuco (Recife, Brazil). The authors have authorization from 4
ACCEPTED MANUSCRIPT the Instituto Chico Mendes de Conservação da Biodiversidade from Brazilian Ministry of the Environment for plant collection (number 38690).
2.2. Lectin preparation
PT
cMoL was obtained in accordance with the protocol described by Luz et al. [13]. Seed
RI
flour proteins were extracted with 0.15 M NaCl at 25°C for 6 h. The extract was treated with
SC
ammonium sulphate (0-60% saturation) at 25°C for 4 h and a precipitated fraction was obtained, dialyzed and applied (10 mg of protein) on a guar gel column (10 cm × 1.0 cm)
NU
previously equilibrated (20 mL/h flow rate) with 0.1 M NaCl. After washing step with 0.15 M NaCl, cMoL was eluted with 0.3 M NaCl. UV absorbance was used to monitor protein
MA
elution. cMoL concentration was evaluated according to Lowry et al. [21] using bovine serum albumin (31.25–500 µg/mL) as standard. Carbohydrate-binding ability was monitored by
PT E
D
hemagglutinating activity assay performed according to Silva et al. [22].
CE
2.3. Cell culture
B16-F10 cells were obtained from American Type Culture Collection (Virginia, USA)
AC
and cultured in 75-cm2 plastic bottles (TPP, Trasadingen, Switzerland) in RPMI 1640 (Vitrocell, São Paulo, Brazil) supplemented with 10% fetal bovine serum (FBS), 100 µg/mL gentamicin, 100 IU/mL penicillin and 100 µg/mL streptomycin (Vitrocell, São Paulo, Brazil). Primary GN fibroblasts were obtained from normal gingival biopsy and kindly provided by Dr. Ricardo Della Coletta from the Universidade Estadual de Campinas. GN cells were grown in DMEM with high glucose supplemented with 10% FBS (Vitrocell, São Paulo,
5
ACCEPTED MANUSCRIPT Brazil), 2 mM glutamine, 100 µg/mL gentamicin, 100 IU/mL penicillin, and 100 µg/mL streptomycin. The cells were kept at 37°C in a humidified atmosphere with 5% CO2.
2.4. cMoL effects on B16-F10 cell viability
PT
The effect of cMoL on cell viability was evaluated as previously described [23].
RI
Briefly, B16-F10 cells were plated in 6-well culture plates (3.5 x 104 cells per well) and after
SC
24 h, the medium was replaced and cells were treated with the lectin (1.5–16 µM). The concentrations were calculated considering a native molecular mass of 30 kDa for cMoL [10].
NU
After further 48 h, cells were washed in PBS, treated with a trypsin-EDTA solution (Vitrocell, São Paulo, Brazil), centrifuged at 1500 g for 4 min and resuspended in 300 µL of medium
MA
containing FBS. Cell viability was assessed using trypan blue dye (0.1%, w/v) added to aliquots of cell suspensions, and the percentages of stained (unviable) cells were determined
D
microscopically. Cell viability in lectin treatments was expressed as the percentage of the
CE
control was above 95%.
PT E
number of viable cells in control. The assay was considered valid only when cell viability in
AC
2.5. Flow cytometry analysis of cell death
Samples were analyzed in a FACSCalibur flow cytometer (BD Biosciences, USA) equipped with an argon laser and Cell-Quest software (version 4.1). For the cell death analysis, B16-F10 cells (106) from control or treated with cMoL (1.5–16 µM; 48 h) were incubated in labeling buffer (10 mM HEPES, pH 7.4, 150 mM NaCl, 5 mM KCl, 1 mM MgCl2 and 1.8 mM CaCl2) containing annexin V (Anx V) conjugated to FITC (1:500) and propidium iodide (PI; 20 µg/mL) at room temperature for 20 min in the dark [23]. Ten 6
ACCEPTED MANUSCRIPT thousand events were acquired for each sample and apoptosis was quantified as the percentage of Anx Vpos/PIneg while necrosis was quantified as the number of Anx Vneg/PIpos cells. GN cells treated or not with cMoL (1.5–16 µM; 48 h) were also evaluated for cell death.
PT
2.6. Microscopy
RI
B16-F10 melanoma cells (3.5 × 104 cells/mL) were treated with cMoL at 8 or 16 µM
SC
for 48 h. Then, the cells were photographed under a microscope Leica DFC360 FX with an
NU
increase of 20 x, using LAS AF software (Leica Microsystems, Germany).
MA
2.7. Determination of mitochondrial superoxide generation
B16-F10 cells (106) were treated with cMoL (8 µM) for 6 h, harvested, and stained
D
with 5 µM MitoSOX (Molecular Probes, USA) for 10 min at 37°C in order to detect
PT E
mitochondrial superoxide levels. MitoSOX fluorescence intensity was analyzed by flow
CE
cytometry (FACSCalibur, BD, San Jose, USA).
AC
2.8. Detection of caspases 3, 8 and 9 activation
To determine caspases 3, 8 and 9 activation, B16-F10 cells were treated with cMoL at 8 µM and, after 48 h, 106 cells were stained with the fluorescent markers FITC-DEVD-FMK, FITC-IETD-FMK or FITC-LEHD-FMK (1:300, Calbiochem, USA) in serum-free medium for 40 min at 37°C in a humidified atmosphere with 5% CO2. Then, cells were washed, resuspended in the same medium, and analyzed by flow cytometry according to the manufacturer’s instructions. B16-F10 cells treated with 1.25 µg/mL cycloheximide (Sigma7
ACCEPTED MANUSCRIPT Aldrich, USA) and 10 nM tumor necrosis factor α (TNFα; Peprotech, USA) for 24 h were used as a positive controls.
2.9. Statistical analysis
PT
Data were expressed as the mean ± the standard error of the mean (S.E.M.) of at least
RI
five independent experiments. Differences between means values were analyzed using one-
SC
way ANOVA followed by Tukey’s multi-comparison test. A p value < 0.05 was considered significant. The concentration of cMoL that causes 50% death of melanoma cells (IC 50) was
NU
calculated by probit analysis using the software Statplus LE (AnalystSoft, Canada).
MA
3. Results
PT E
D
3.1. cMoL reduces cell viability and causes cell death on B16-F10 melanoma
cMoL was successfully isolated following the previous published protocol and showed
CE
hemagglutinating activity (256-1), revealing that its ability to bind carbohydrates was preserved. In the present study, the effect of cMoL on the viability and death of B16-F10 cells
AC
was followed using trypan blue exclusion assay and flow cytometry, respectively, after 48 h of exposure in culture. The treatment with increasing concentrations of cMoL progressively reduced cell viability (Figure 1) and caused cell death (Figure 2A). IC50 value was estimated to be 9.72 µM. To characterize the cell death, the rates of apoptosis and necrosis were analyzed by annexin V and PI staining. Figure 2A shows that majority of damaged cells in treatments with cMoL was stained with PI but a considerable subpopulation of PIpos cells was also positive for 8
ACCEPTED MANUSCRIPT annexin V staining. Since cMoL at 8 µM induced 47.6% ± S.E.M. cell death in B16-F10 cells, the treatment at this concentration was established for the following experiments. The effect of cMoL on normal fibroblasts (GN) was also analyzed. In contrast to results obtained with B16-F10 melanoma cells, cMoL did not cause death of more than 20% of GN cells after 48 h (Figure 2B). Both apoptotic and necrotic cells were detected.
PT
Marked morphologic alterations were observed by optical microscopy in B16-F10
RI
cells cultured in the presence of cMoL at 8 and 16 µM for 48 h (Figure 2C). Progressive
SC
decrease in cell number and morphological changes as rounded cells and reduction of cellular
NU
volume were observed.
MA
3.2. cMoL induces caspase activation and ROS production by B16-F10 cell line
Activation of caspases 3, 8 and 9 was detected in B16-F10 cells treated with cMoL
D
(Figure 3), suggesting that this lectin induces apoptotic cell death in a caspase-dependent
PT E
manner. Positive controls cycloheximide (1.25 µg/mL) and TNF-α (10 nM) promoted caspase 8 activation as expected. The evaluation of ROS production revealed that, after 6 h, cMoL
AC
4. Discussion
CE
promoted a two-fold increase of mitochondrial ROS level (Figure 4).
The results obtained in the present work show that cMoL causes cell death on B16F10 melanoma and was less cytotoxic to GN cells. This finding suggests that this lectin bears a greater recognition for malignant cells. Other lectins have demonstrated to be active on cancer cells with low toxicity to normal cells. For example, the Microgramma vacciniifolia rhizome lectin was toxic to lung carcinoma cell NCI-H292 cells (IC50 = 25.23 µg/mL) but did 9
ACCEPTED MANUSCRIPT not reduce the viability of human peripheral blood mononuclear cells (PBMCs) even when tested at 100 µg/mL [24]. Similarly, the lectin from Calliandra surinamensis leaf pinnulae reduced the viability of leukemic K562 cells (IC50 = 67.04 µg/mL) and breast cancer T47D cells (IC50 = 58.75 µg/ mL) without any cytotoxic effect on PBMCs [25]. This behavior can be explained by the binding specificity of the lectin regarding the differential expression of
PT
carbohydrates at the plasma membranes of normal and malignant cells. When the lectin is
RI
able to bind plasma membrane receptors and/or is internalized by endocytosis, it may trigger a
SC
variety of cellular events such as apoptosis, autophagy, necrosis or mitotic catastrophe [26]. Raz et al. [27] studied the composition of surfaces of melanoma cells, including B16-
NU
F10. They reported evidences of the presence of galactose and sialic acid and investigation through lectin binding showed that the B16-F10 surface was recognized by concanavalin A
MA
(glucose/mannose binding protein), soybean agglutinin (that recognizes galactose) and wheat germ agglutinin (N-acetylglucosamine-binding lectin). Santos et al. [10] showed that cMoL
D
has higher affinity for the monosaccharides glucose and galactose. In addition, they proved
PT E
that this lectin is able to recognize not only monosaccharides but also sugars present in the oligosaccharide moieties of glycoproteins. In this sense, cMoL may be recognizing glucose
CE
and galactose residues present in glycoconjugates found at B16-F10 cells surface and thus trigger intracellular events leading to cell death.
AC
Necrosis can be attributed as important cell death type induced by cMoL on B16-F10 cells due to the remarkable staining with PI. In addition, the presence of double staining for both annexin V and PI suggests cells in late apoptosis or necrotic cells already dead [28]. Annexin V can stain cells in advanced necrotic stage because it may enter the cell through the ruptured membranes and then access phosphatidylserine at the inner layer of the plasma membrane [29]. To shed some light on the death mechanisms promoted by cMoL, we
10
ACCEPTED MANUSCRIPT investigated a hypothesis that part of the B16-F10 cells treated with the lectin entered in an apoptosis-related pathway by assessing the activation of caspases. Indeed, the activation of caspase-3 confirms apoptotic cell death induced by cMoL in B16-F10 cells and the activation of caspases 8 and 9 reflects the involvement of both extrinsic and intrinsic signaling pathways, respectively. The cascades involved in extrinsic and intrinsic
PT
pathways are different but both can be merged into caspase activation. To initiate the extrinsic
RI
pathway, it is necessary an extracellular stimuli such as UV, X-ray or a toxin. Then, the
SC
combination between Fas-L (Fas ligand) and Fas (membrane receptor) forms a death complex and results in the intracellular activation of initiator caspase-8 and executioner caspases 3 and
NU
6 [30]. cMoL should act as a potent stimulus for activation of extrinsic pathway. The activation of caspase-3 is involved in the cell death mechanism described for
MA
other lectins that induce apoptosis. The Polygonatum odoratum lectin is able to induce human lung adenocarcinoma A549 cells apoptosis with activation of caspase-3 mediated by the
D
microRNA miR-15a-3p [31]. A lectin from the hemolymph of the millipede Gluttonous
PT E
beauts induced apoptosis in MCF-7 cells through up-regulation of Bad and caspase-3 expression [32]. Similarly to cMoL, the lectin from Rhizoctonia bataticola activated extrinsic
CE
apoptotic pathway mediated by caspase-8 in leukemic cells Molt-4 and Jurkat; the authors also reported that this lectin was not active on normal lymphocytes [33].
AC
Numerous pro-apoptotic signals are stimulated by several factors, among them, increased ROS production, which in turn characterizes the condition of oxidative stress and can precede cell death [16,34]. Thus, we also evaluated whether cMoL stimulated the ROS production by mitochondria of B16-F10 cells, which was confirmed by the results obtained. The increased levels of ROS may result in oxidation of amino acids, polydesaturated fatty acids as well as cause DNA damage, leading cell to death [35]. A pro-oxidant status induced by Aronia melanocarpa juice is involved in the apoptosis induced in Jurkat cells [36] and the 11
ACCEPTED MANUSCRIPT Polygonatum cyrtonema lectin induces apoptosis and autophagy of melanoma A375 cells inducing the massive ROS accumulation in mitochondria [37]. Apoptosis is accompanied by cell rounding-up, retraction of pseudopodes, reduction of cellular volume (pyknosis), chromatin condensation, nuclear fragmentation (karyorrhexis), little or no ultrastructural modifications of cytoplasmic organelles, and plasma membrane
RI
cMoL also induced apoptotic death of B16-F10 melanoma cells.
PT
blebbing [38]. Some of the results obtained in microscopy analysis support the possibility that
SC
Although apoptosis and necrosis have often been treated as excluding events, it is now known that they have common sub-cellular sites and organelles, may share initiator and
NU
effector molecules and can co-operate in a balanced interplay [39]. Interesting, our results show that part of B16-F10 cells entered in a necrotic process when incubated with cMoL but
MA
we also found evidences that some cells also underwent apoptotic process. These findings stimulate future studies focusing on the evaluation of deeper molecular mechanisms aiming at
D
understanding in detail the action of cMoL on B16-F10 cell death.
PT E
Exogenous glycan-binding proteins (e.g. plant lectins) may establish multivalent interactions with transmembrane glycoproteins and transduce intracellular signals leading to
CE
cell death. The interaction of lectin with glycosylated cell death receptors may modulate positively or negatively the apoptotic machinery. These interactions may also affect signaling
AC
thresholds and receptor clustering as well as lead to lectin endocytosis [40]. The lectin from Artocarpus heterophyllus seeds was cytotoxic to NB4 (human myelocytic leukemia) cells strongly inducing reactive oxygen species generation and cell death was attributed to recognition by the lectin of a trimannosyl core of N-glycans containing a β1,6-GlcNAc branch linked to α1,6-mannose [41]. The cMoL may exert its cytotoxic effects on B16-F10 cells leading to drastic deregulation of cell homeostasis (resulting in necrosis) and triggering caspase activation and ROS production pathways. The initiation of these processes may 12
ACCEPTED MANUSCRIPT include internalization by the cell or interaction with glycosylated receptors, which remain to be defined in the future.
5. Conclusions
PT
The lectin cMoL showed cytotoxic effects on B16-F10 melanoma cells, with
RI
specificity for these cancer cells compared to normal human fibroblasts (GN). The study
SC
stimulates more investigation on the anticancer potential and possible therapeutic uses of
NU
cMoL.
MA
Conflict of Interest statement
PT E
Acknowledgements
D
The authors declare that there are no conflicts of interest.
CE
We are thankful to Dr. Anibal Eugênio Vercesi from the Departamento de Patologia Clínica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas (UNICAMP)
AC
for support and scientific assistance. We are also grateful to Dr. Carlos Amilcar Parada and Raffaela Silvestre Ignarro, from UNICAMP, for assistance in optical microscopy. The authors express their gratitude to the Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP; 11/50400-0), to the Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) for research grants (446902/2014-4) and fellowships (THN, PMGP and LCBBC). The Fundação de Amparo à Ciência e Tecnologia do Estado de Pernambuco
13
ACCEPTED MANUSCRIPT (FACEPE; APQ-0661-2.08/15) and the Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES) for research grants and financial support are also acknowledged.
References
PT
[1] Teixeira CMLL, Kirsten FV, Teixeira PCN. Evaluation of Moringa oleifera seed flour as
RI
a flocculating agent for potential biodiesel producer microalgae. J Appl Phycol 2012;24:557–
SC
63.
[2] Santos AFS, Luz LA, Pontual EV, Napoleão TH, Paiva PMG, Coelho LCBB. Moringa
NU
oleifera: Resource management and multiuse life tree. Adv Res 2015;4:388-402. [3] Cheenpracha S, Park E-J, Yoshida WY, Barit C, Wall M, Pezzuto JM, Chang LC.
MA
Potential anti-inflammatory phenolic glycosides from the medicinal plant Moringa oleifera fruits. Bioorg Med Chem 2010;18:6598–602.
D
[4] Araújo LCC, Aguiar JS, Napoleão TH, Mota FVB, Barros ALS, Moura MC, Coriolano
PT E
MC, Coelho LCBB, Silva TG, Paiva PMG. Evaluation of cytotoxic and anti-inflammatory activities of extracts and lectins from Moringa oleifera seeds. PLoS ONE 2013;8:e81973.
CE
[5] Choudhary MK, Bodakhe SH, Gupta SK. Assessment of the antiulcer potential of Moringa oleifera root-bark extract in rats. J Acupunct Meridian Stud 2013;6:214-20.
AC
[6] Jung IL. Soluble extract from Moringa oleifera leaves with a new anticancer activity. PLoS ONE 2014;9:e95492. [7] Al-Asmari AK, Albalawi SM, Athar MT, Khan AQ, Al-Shahrani H, Islam M. Moringa oleifera as an anti-cancer agent against breast and colorectal cancer cell lines. PLoS ONE 2015;10:e0135814. [8] Ingale SP, Gandhi FP. Effect of aqueous extract of Moringa oleifera leaves on pharmacological models of epilepsy and anxiety in mice. Int J Epilepsy 2016;3:12-9. 14
ACCEPTED MANUSCRIPT [9] Metwally FM, Rashas HM, Ahmed HH, Mahmoud AA, Raouf ERA, Abdalla AM. Molecular mechanisms of the anti-obesity potential effect of Moringa oleifera in the experimental model. Asian Pac J Trop Biomed 2017;doi:10.1016/j.apjtb.2016.12.007 [10] Santos AFS, Luz LA, Argolo ACC, Teixeira JA, Paiva PMG, Coelho LCBB. Isolation of a seed coagulant Moringa oleifera lectin. Process Biochem 2009;44:504–8.
PT
[11] Ferreira RS, Napoleão TH, Santos AFS, Sá RA, Carneiro-da-Cunha MG, Morais MMC,
RI
Silva-Lucca RA, Oliva MLV, Coelho LCBB, Paiva PMG. Coagulant and antibacterial
SC
activities of the water-soluble seed lectin from Moringa oleifera. Lett Appl Microbiol 2011;53:186-92.
NU
[12] Procópio TF, Moura MC, Albuquerque LP, Gomes FS, Santos NDL, Coelho LCBB, Pontual EV, Paiva PMG, Napoleão TH. Antibacterial Lectins: Action Mechanisms, Defensive
MA
Roles and Biotechnological Potential. In: Collins, E. (Ed.) Antibacterials: Synthesis, Properties and Biological Activities. 1st ed. New York: Nova Science Publishers Inc.; 2016
D
[chapter 3]
PT E
[13] Luz LA, Silva MCC, Ferreira RS, Santana LA, Silva-Lucca RA, Mentele R, Oliva MLV, Paiva PMG, Coelho LCBB. Structural characterization of coagulant Moringa oleifera lectin
CE
and its effect on hemostatic parameters. Int J Biol Macromol 2013;58:31-6. [14] Oliveira CFR, Luz LA, Paiva PMG, Coelho LCBB, Marangoni S, Macedo MLR.
AC
Evaluation of seed coagulant Moringa oleifera lectin (cMoL) as a bioinsecticidal tool with potential for the control of insects. Process Biochem 2011;46:498–504. [15] Fulda S, Kroemer G. Mitochondria as therapeutic targets for the treatment of malignant disease. Antioxid Redox Signal 2011;15:2937-49. [16] Figueira TR, Barros MH, Camargo AA, Castilho RF, Ferreira JC, Kowaltowski AJ, Sluse FE, Souza-Pinto NC, Vercesi AE. Mitochondria as a source of reactive oxygen and
15
ACCEPTED MANUSCRIPT nitrogen species: from molecular mechanisms to human health. Antioxid Redox Signal 2013;18:2029–74. [17] Fernandes MP, Inada NM, Chiaratti MR, Araújo FF, Meirelles FV, Correia MT, Coelho LC, Alves MJ, Gadelha FR, Vercesi AE. Mechanism of Trypanosoma cruzi death induced by Cratylia mollis seed lectin. J Bioenerg Biomembr 2010;42:69-78.
PT
[18] Osterne VJS, Silva-Filho JC, Santiago MQ, Pinto-Junior VR, Almeida AC, Barreto
RI
AAGC, Wolin IAV, Nascimento APM, Amorim RMF, Rocha BAM, Delatorre P, Nagano
SC
CS, Leal RB, Assreuy AMS, Nascimento KS, Cavada BS. Structural characterization of a lectin from Canavalia virosa seeds with inflammatory and cytotoxic activities. Int J Biol
NU
Macromol 2017;94:271–82.
[19] Damasio, DC, Nolte S, Polak LP, Brandt AP, Bonan NB, Zischler L, Stuelp-Campelo
MA
PM, Cadena SMSC, Noronha L, Elífio-Esposito SL, Moreno-Amaral AN. The lectin BJcuL induces apoptosis through TRAIL expression, caspase cascade activation and mitochondrial
D
membrane permeability in a human colon adenocarcinoma cell line. Toxicon 2014;90:299-
PT E
307.
[20] Zang G, Gustafsson K, Jamalpour M, Hing JW, Genové G, Welsh M. Vascular
CE
dysfunction and increased metastasis of B16F10 melanomas in Shb deficient mice as compared with their wild type counterparts. BMC Cancer 2015;15:234.
AC
[21] Lowry OH, Rosebrough NJ, Farr AL, Randall RJ. Protein measurement with the folin phenol reagent. J Biol Chem 1951;193:265–75. [22] Silva PM, Napoleão TH, Silva LCPBB, Fortes DTO, Lima TA, Zingali RB, Pontual EV, Araujo JM, Medeiros PL, Rodrigues CG, Gomes FS, Paiva PMG. The juicy sarcotesta of Punica granatum contains a lectin that affects growth, survival as well as adherence and invasive capacities of human pathogenic bacteria. J Funct Foods 2016;27:695-702.
16
ACCEPTED MANUSCRIPT [23] Rossato FA, Zecchin KG, La Guardia PG, Ortega RM, Alberici LC, Costa RA, Catharino RR, Graner E, Castilho RF, Vercesi AE. Fatty Acid synthase inhibitors induce apoptosis in non-tumorigenic melan-a cells associated with inhibition of mitochondrial respiration. PLoS ONE 2014;9:e101060. [24] Albuquerque LP, Pontual EV, Santana GMS, Silva LRS, Aguiar JS, Coelho LCBB, Rêgo
PT
MJBM, Pitta MGR, Silva TG, Melo AMMA, Napoleão TH, Paiva PMG. Toxic effects of
RI
Microgramma vacciniifolia rhizome lectin on Artemia salina, human cells, and the
SC
schistosomiasis vector Biomphalaria glabrata. Acta Trop 2014;138:23–7. [25] Procópio TF, Patriota LLS, Moura MC, Silva PM, Oliveira APS, Carvalho LVN, Lima
NU
TA, Soares T, Silva TD, Coelho LCBB, Pitta MGR, Rêgo MJBM, Figueiredo RCBQ, Paiva PMG, Napoleão TH. CasuL: A new lectin isolated from Calliandra surinamensis leafpinnulae
MA
with cytotoxicity to cancer cells, antimicrobial activity and antibiofilm effect. Int J Biol Macromol 2017;98:419-429.
D
[26] Galluzzi L, Vitale I, Vacchelli E, Kroemer G. Cell death signaling and anticancer
PT E
therapy. Front Oncol. 2011;1:1-19.
[27] Raz A, McLellan WL, Hart IR, Bucana CD, Hoyer LC, Sela B-A., Dragsten P, Fidler.
CE
Cell surface properties of B16 melanoma variants with differing metastatic potential. Cancer Res 1980;40:1645-1651.
AC
[28] Hingonari R, Deng J, Elia J, McIntyre C, Mitar D. Detection of apoptosis using the BD Annexin V FITC Assay on the BD FACSVerse™ system. Application Note. San Jose: BD Biosciences, 2011. [29] Crowley LC, Marfell BJ, Scott AP, Waterhouse NJ. Quantitation of apoptosis and necrosis by Annexin V binding, propidium iodide uptake, and flow cytometry. Cold Spring Harb Protoc 2016;2016:pdb.prot087288.
17
ACCEPTED MANUSCRIPT [30] Lavrik I, Golks A, Krammer PH. Death receptor signaling. J Cell Science 2005;118:2657. [31] Wu L, Liu T, Xiao Y, Li X, Zhu Y, Zhao Y, Bao J, Wu C. Polygonatum odoratum lectin induces apoptosis and autophagy by regulation of microRNA-1290 and microRNA-15a-3p in human lung adenocarcinoma A549 cells. Int J Biol Macromol 2016;85:217-26.
PT
[32] Ponraj T, Paulpandi M, Vivek R, Vimala K, Kanna S. Protein regulation and Apoptotic
RI
induction in human breast carcinoma cells (MCF-7) through lectin from G. beauts. Int J Biol
SC
Macromol 2017;95:1235-1245.
[33] Pujari R, Eligar SM, Kumar N, Barkeer S, Reddy V, Swamy BM, Inamdar SR, Shastry
NU
P. Rhizoctonia bataticola lectin (RBL) induces caspase-8-mediated apoptosis in human T-cell leukemia cell lines but not in normal CD3 and CD34 positive cells. PLoS ONE
MA
2013;8:e79311.
[34] Kowaltowski AJ, Vercesi AE. Mitochondrial damage induced by conditions of oxidative
D
stress. Free Radic Biol Med 1999;26:463-71.
PT E
[35] Mazure NM, Pouysségur J. Hypoxia-induced autophagy: cell death or cell survival? Cur Opinion in Cell Biol 2010;22:177-180.
CE
[36] Sharif T, Alhosin M, Auger C, Minker C, Kim J-H, Etienne-Selloum N, Bories P, Gronemeyer H, Lobstein A, Bronner C, Fuhrmann G, Schini-Kerth VB. Aronia melanocarpa
AC
juice induces a redox-sensitive p73-related caspase 3-dependent apoptosis in human leukemia cells. PLoS ONE 2012;7:e32526. [37] Liu B, Cheng Y, Zhang B, Bian HJ, Bao JK. Polygonatum cyrtonema lectin induces apoptosis and autophagy in human melanoma A375 cells through a mitochondria-mediated ROS-p38-p53 pathway. Cancer Lett 2009;275:54–60. [38] Kroemer G, Galluzzi L, Vandenabeele P, Abrams J, Alnemri ES, Baehrecke EH, Blagosklonny MV, El-Deiry WS, Golstein P, Green DR, Hengartner M, Knight RA, Kumar 18
ACCEPTED MANUSCRIPT S, Lipton SA, Malorni W, Nunez G, Peter ME,
Tschopp J, Yuan J, Piacentini M,
Zhivotovsky B, Melino G. Classification of cell death: recommendations of the Nomenclature Committee on Cell Death. Cell Death Differ 2009;16:3-11. [39] Nikoletopoulou V, Markaki M, Palikaras K, Tavernarakis N. Crosstalk between apoptosis, necrosis and autophagy. Biochim Biophys Acta 2013;1833:3448–3459.
PT
[40] Lichtenstein RG, Rabinovich GA. Glycobiology of cell death: when glycans and lectins
RI
govern cell fate. Cell Death Differ 2013;20:976-986.
SC
[41] Carvalho FC, Soares SG, Tamarozzi MB, Rego EM, Roque-Barreira M-C. The recognition of N-Glycans by the lectin ArtinM mediates cell death of a human myeloid
NU
leukemia cell line. PLoS ONE 2011;11:e27892.
MA
Figure captions
D
Figure 1. cMoL reduces B16-F10 melanoma cell viability. B16-F10 cells were treated with
PT E
increasing concentrations of the lectin for 48 h. Cell viability was expressed as the percentage of viable cells in regard to control. Cell viability under control condition was above 95%.
CE
Values are mean ± S.E.M. of at least five independent experiments. A p-value < 0.05 was
AC
considered to indicate significance; p-value < 0.05 (*) and p-value < 0.001 (**).
Figure 2. cMoL induces cell death in B16-F10 melanoma cells (A) but not on GN normal fibroblasts (B). Cells were treated with increasing concentrations of cMoL for 48 h, stained with annexin V and propidium iodide. The percentages of necrotic (PI+) or apoptotic (AnxV+) cells were determined by flow cytometry. Values are mean ± S.E.M. of at least five independent experiments. *Significantly different from control at p < 0.05, Student’s t-test.
19
ACCEPTED MANUSCRIPT (C) Representative images of control and cMoL-treated cells provided by a Leica DFC360 FX microscope. Bars represent 100 μm.
Figure 3. cMoL treatment induces caspase 3 (A), 8 (B) and 9 (C) activation. Caspase activation was determined by flow cytometry using FITC-DEVD-FMK after 48 h of treatment
PT
with the lectin. Values are mean ± S.E.M of at least 5 independent experiments; p-value <
SC
RI
0.05 (*) and p-value < 0.001 (**).
Figure 4. Treatment of B16-F10 melanoma cells with cMoL increases mitochondrial
NU
superoxide levels. Cells (106/mL) were treated with cMoL for 6 h, then washed and probed with 5 µM MitoSOX at 37 °C for 10 min. Superoxide levels were analyzed by flow
MA
cytometry. Values are the means ± S.E.M. of at least five independent experiments.
AC
CE
PT E
D
*Significantly different from control at p < 0.05 (*) and p-value < 0.001 (**) level.
20
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
MA
Figure 1
21
MA
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
Figure 2
22
D
MA
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
AC
CE
PT E
Figure 3
23
MA
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
Figure 4
24
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
AC
CE
PT E
D
MA
Graphical abstract
25
ACCEPTED MANUSCRIPT Highlights
The cytotoxicity of the lectin cMoL on B16-F10 murine melanoma cells was evaluated. cMoL reduced viability of B16-F10 cells and was low toxic to normal human
PT
fibroblasts.
RI
Flow cytometry analysis revealed necrotic and late apoptotic cells
SC
Rounded shape and volume reduction were observed in B16-F10 cells treated with cMoL.
AC
CE
PT E
D
MA
NU
cMoL increased mitochondrial ROS and activated caspases 3, 8 and 9 in B16-F10.
26