Acessibilidade / Reportar erro

Cytotoxic effects of crotoxin from Crotalus durissus terrificus snake in canine mammary tumor cell lines

Abstract

Background:

Mammary gland tumors are the most prevalent neoplasm in intact female dogs, and they are good natural models to study comparative oncology. Most canine mammary malignancies, as in women, are commonly refractory to conventional therapies and demand continuous new therapeutic approaches. Crotalus durissus terrificus, also called rattlesnake, has more than 60 different proteins in its venom with multiple pharmaceutical uses, such as antitumor, antiviral, and antimicrobial action. Crotoxin, a potent β-neurotoxin formed by the junction of two subunits, a basic subunit (CB-PLA2) and an acidic subunit (crotapotin), has already been reported to have anticancer properties in different types of cancers.

Methods:

In this work, we describe the cytotoxic potential of crotoxin and its subunits compared to doxorubicin (drug of choice) in two canine mammary carcinoma cell lines.

Results:

Crotoxin, CB-PLA2, crotalic venom, and doxorubicin decreased cell viability and the ability to migrate in a dose-dependent manner, and crotapotin did not present an antitumoral effect. For all compounds, the predominant cell death mechanism was apoptosis. In addition, crotoxin did not show toxicity in normal canine mammary gland cells.

Conclusion:

Therefore, this work showed that crotoxin and CB-PLA2 had cytotoxic activity, migration inhibition, and pro-apoptotic potential in canine mammary gland carcinoma cell lines, making their possible use in cancer research.

Keywords:
Mammary tumor; Comparative oncology; Crotalus durissus terrificus venom; Biological compounds

Background

Mammary gland tumors are the most prevalent neoplasm in intact female dogs, but the incidence can vary and be as high as 83% in a study population [11. Nakagaki KY, Nunes MM, Garcia APV, Nunes FC, Schmitt F, Cassali GD. Solid Carcinoma of the Canine Mammary Gland: a Histological Type or Tumour Cell Arrangement? J Comp Pathol. 2022 Jan;190:1-12., 22. Nunes FC, Campos CB, Teixeira SV, Bertagnolli AC, Lavalle GE, Cassali GD. Epidemiological, clinical and pathological evaluation of overall survival in canines with mammary neoplasms. Arq Bras Med Vet Zootec. 2018 Nov-Dez;70(6):1714-22.]. Comparatively, in 2022, Brazil’s public health system reported 73,610 new diagnoses of breast cancer in women, 30.1% of all newly diagnosed neoplasias, which is more than three times what ranked second in statistics [33. Instituto Nacional de Cancer José Alencar Gomes Silva. Estimativa 2023: incidência do Câncer no Brasil. Rio de Janeiro: INCA, 2022. Disponível em: https://www.gov.br/inca/ptbr/assuntos/cancer/numeros/estimativa.
https://www.gov.br/inca/ptbr/assuntos/ca...
].

Canine cancers occur spontaneously and have a similar clinical presentation and pathophysiology to humans, in addition to sharing the main risk factors and signaling pathways [44. Kwon JY, Moskwa N, Kang W, Fan TM, Lee C. Canine as a Comparative and Translational Model for Human Mammary Tumor. J Breast Cancer. 2023 Feb;26(1):1-13., 55. Raffo-Romero A, Aboulouard S, Bouchaert E, Rybicka A, Tierny D, Hajjaji N, Fournier I, Salzet M, Duhamel M. Establishment and characterization of canine mammary tumoroids for translational research. BMC Biol. 2023 Feb 3;21(1):23.], such as the expression of the HER-2 protein and Ki-67, related to tumor aggressiveness. This makes dogs a valuable study model as they are more similar to the natural progression of human cancer than induced animal cancer models [66. Antuofermo E, Miller MA, Pirino S, Xie J, Badve S, Mohammed SI. Spontaneous mammary intraepithelial lesions in dogs--a model of breast cancer. Cancer Epidemiol Biomarkers Prev. 2007 Nov;16(11):2247-56.]. Most canine mammary malignancies are commonly refractory to conventional therapies and demand continuous new therapeutic approaches, as in women. Biological compounds are the source of 25% of newly approved medicines used in cancer treatment in recent decades in natural or re-engineered structures [77. Huang M, Lu JJ, Ding J. Natural Products in Cancer Therapy: Past, Present and Future. Nat Prod Bioprospect. 2021;11:5-13., 88. Newman DJ, Cragg GM. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019. J Nat Prod. 2020 Mar 27;83(3):770-803.].

As a source of multiple biological molecules, snake venom is a tremendous opportunity for new medical uses by improvement with a biotechnological process [99. Alves BFA, Ferreira RS Jr. Antineoplastic properties and pharmacological applications of Crotalus durissus terrificus snake venom. Rev Soc Bras Med Trop. 2022 Dec 16;55:e0323-2022., 1010. Abbade LPF, Barraviera SRCS, Silvares MRC, Lima ABBCO, Haddad GR, Gatti MAN, Medolago NB, Rigotto Carneiro MT, Dos Santos LD, Ferreira RS Jr, Barraviera B. Treatment of Chronic Venous Ulcers With Heterologous Fibrin Sealant: A Phase I/II Clinical Trial. Front Immunol. 2021 Feb 23;12:627541., 1111. Diniz-Sousa R, Caldeira CADS, Pereira SS, Da Silva SL, Fernandes PA, Teixeira LMC, Zuliani JP, Soares AM. Therapeutic applications of snake venoms: An invaluable potential of new drug candidates. Int J Biol Macromol. 2023 May 31;238:124357., 1212. Mohamed Abd El-Aziz T, Garcia Soares A, Stockand JD. Snake Venoms in Drug Discovery: Valuable Therapeutic Tools for Life Saving. Toxins (Basel). 2019 Sep 25;11(10):564., 1313. Oliveira AL, Viegas MF, da Silva SL, Soares AM, Ramos MJ, Fernandes PA. The chemistry of snake venom and its medicinal potential. Nat Rev Chem. 2022 Jun 10;6:451-69., 1414. Shahbazi B, Najafabadi ZS, Goudarzi H, Sajadi M, Tahoori F, Bagheri M. Cytotoxic effects of Pseudocerastes persicus venom and its HPLC fractions on lung cancer cells. J Venom Anim Toxins incl Trop Dis. 2019 Sep 16;25:e20190009. doi: 10.1590/1678-9199-JVATITD-2019-0009. PMID: 31555336; PMCID: PMC6748451.
https://doi.org/10.1590/1678-9199-JVATIT...
]. Crotalus durissus terrificus, a Viperidae snake (Crotalinae subfamily) also called Rattlesnake, and its venom (CdtV) comprises more than 60 different proteins with multiple pharmaceutical uses, such as antitumor, antiviral, and antimicrobial activities [99. Alves BFA, Ferreira RS Jr. Antineoplastic properties and pharmacological applications of Crotalus durissus terrificus snake venom. Rev Soc Bras Med Trop. 2022 Dec 16;55:e0323-2022., 1515. Calderon LA, Sobrinho JC, Zaqueo KD, de Moura AA, Grabner AN, Mazzi MV, Marcussi S, Nomizo A, Fernandes CF, Zuliani JP, Carvalho BM, da Silva SL, Stábeli RG, Soares AM. Antitumoral activity of snake venom proteins: new trends in cancer therapy. Biomed Res Int. 2014;2014:203639., 1616. Macedo JM, Lima AM, Kayano AM, Souza MF, Oliveira IS, Garay AFG, Rocha AM, Zuliani JP, Soares AM. Literature review on Crotalus durissus terrificus toxins: From a perspective of structural biology and therapeutic applications. Curr Protein Pept Sci. 2023;24(7):536-50.]. Crotoxin (CTX) is a potent β-neurotoxin formed by the junction of two subunits: one basic (CB-PLA2) enzymatic part and another acidic (crotapotin) with structural properties, exhibits nephrotoxic, cardiotoxic, and myotoxic effects [1717. Sartim MA, Menaldo DL, Sampaio SV. Immunotherapeutic potential of Crotoxin: anti-inflammatory and immunosuppressive properties. J Venom Anim Toxins incl Trop Dis. 2018 Dec 17;24:39. doi: 10.1186/s40409-018-0178-3. PMID: 30564276; PMCID: PMC6296157.
https://doi.org/10.1186/s40409-018-0178-...
] and this complex protein represents approximately 50% of dry crude venom [99. Alves BFA, Ferreira RS Jr. Antineoplastic properties and pharmacological applications of Crotalus durissus terrificus snake venom. Rev Soc Bras Med Trop. 2022 Dec 16;55:e0323-2022., 1212. Mohamed Abd El-Aziz T, Garcia Soares A, Stockand JD. Snake Venoms in Drug Discovery: Valuable Therapeutic Tools for Life Saving. Toxins (Basel). 2019 Sep 25;11(10):564., 1818. Almeida CF, Amaral C, Augusto TV, Correia-da-Silva G, Marques de Andrade C, Torqueti MR, Teixeira N. The anti-cancer potential of crotoxin in estrogen receptor-positive breast cancer: Its effects and mechanism of action. Toxicon. 2021 Sep;200:69-77., 1919. de Oliveira LA, Ferreira RS Jr, Barraviera B, de Carvalho FCT, de Barros LC, Dos Santos LD, Pimenta DC. Crotalus durissus terrificus crotapotin naturally displays preferred positions for amino acid substitutions. J Venom Anim Toxins incl Trop Dis. 2017 Nov 28;23:46. doi: 10.1186/s40409-017-0136-5. PMID: 29209361; PMCID: PMC5704381.
https://doi.org/10.1186/s40409-017-0136-...
, 2020. Sampaio SC, Hyslop S, Fontes MR, Prado-Franceschi J, Zambelli VO, Magro AJ, Brigatte P, Gutierrez VP, Cury Y. Crotoxin: novel activities for a classic beta-neurotoxin. Toxicon. 2010 Jun 1;55(6):1045-60., 2121. Fortes-Dias CL, Lin Y, Ewell J, Diniz CR, Liu TY. A phospholipase A2 inhibitor from the plasma of the South American rattlesnake ( Crotalus durissus terrificus). Protein structure, genomic structure, and mechanism of action. J Biol Chem. 1994 Jun 3;269(22):15646-51.].

In recent decades, CTX has been the most analyzed toxin derived from crude snake venom and used as a multimodal natural agent, with functions such as immunomodulatory, antimicrobial, anticancer, anti-inflammatory, and analgesic compounds [2020. Sampaio SC, Hyslop S, Fontes MR, Prado-Franceschi J, Zambelli VO, Magro AJ, Brigatte P, Gutierrez VP, Cury Y. Crotoxin: novel activities for a classic beta-neurotoxin. Toxicon. 2010 Jun 1;55(6):1045-60., 2222. Sant’Anna MB, Lopes FSR, Kimura LF, Giardini AC, Sant’Anna OA, Picolo G. Crotoxin Conjugated to SBA-15 Nanostructured Mesoporous Silica Induces Long-Last Analgesic Effect in the Neuropathic Pain Model in Mice. Toxins (Basel). 2019 Nov 20;11(12):679., 2323. Teixeira NB, Sant’Anna MB, Giardini AC, Araujo LP, Fonseca LA, Basso AS, Cury Y, Picolo G. Crotoxin down-modulates pro-inflammatory cells and alleviates pain on the MOG35-55-induced experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis. Brain Behav Immun. 2020 Feb;84:253-68., 2424. Sartim MA, Menaldo DL, Sampaio SV. Immunotherapeutic potential of Crotoxin: anti-inflammatory and immunosuppressive properties. J Venom Anim Toxins incl Trop Dis. 2018;24:39. https://doi.org/10.1186/s40409-018-0178-3
https://doi.org/10.1186/s40409-018-0178-...
, 2525. Wolz-Richter S, Esser KH, Hess A. Antinociceptive activity of crotoxin in the central nervous system: a functional Magnetic Resonance Imaging study. Toxicon. 2013 Nov;74:44-55.]. The ability to regulate cytotoxin production in the microenvironment, modulate cell proliferation signaling, promote cell cycle arrest or induction of apoptosis/autophagy, and specifically activate/deactivate cell membrane receptors are the main mechanisms of action in cancer cells [1818. Almeida CF, Amaral C, Augusto TV, Correia-da-Silva G, Marques de Andrade C, Torqueti MR, Teixeira N. The anti-cancer potential of crotoxin in estrogen receptor-positive breast cancer: Its effects and mechanism of action. Toxicon. 2021 Sep;200:69-77., 2626. Almeida TC, Ribeiro Silva LM, Boaventura de Oliveira AM, Lopes FSR, Sant’Anna MB, Picolo G. Cytotoxic effect of crotoxin on cancer cells and its antitumoral effects correlated to tumor microenvironment: A review. Int J Biol Macromol. 2023 May 15;242(Pt 2):124892.].

This neurotoxin has been studied in vitro, in vivo, and more recently in clinical trials, alone or in combination, to develop new human medicines [2727. Cura JE, Blanzaco DP, Brisson C, Cura MA, Cabrol R, Larrateguy L, Mendez C, Sechi JC, Silveira JS, Theiller E, de Roodt AR, Vidal JC. Phase I and pharmacokinetics study of crotoxin (cytotoxic PLA(2), NSC-624244) in patients with advanced cancer. Clin Cancer Res. 2002 Apr;8(4):1033-41., 2828. Medioni J, Brizard M, Elaidi R, Reid PF, Benlhassan K, Bray D. Innovative design for a phase 1 trial with intra-patient dose escalation: The Crotoxin study. Contemp Clin Trials Commun. 2017 Jul 23;7:186-8., 2929. Wang J, Qin X, Zhang Z, Chen M, Wang Y, Gao B. Crotoxin suppresses the tumorigenic properties and enhances the antitumor activity of Iressa® (gefinitib) in human lung adenocarcinoma SPCA-1 cells. Mol Med Rep. 2014 Dec;10(6):3009-14.]. In canine models, this promisor agent, to the best of our knowledge, has not yet been reported. This study aimed to describe the cytotoxic potential and migration inhibition capacity of crotoxin and its subunits compared to doxorubicin (drug of choice) in two canine mammary carcinoma cell lines.

Methods

Materials and chemicals

The reagents used were: antibiotic antimycotic - solution (Gibco™, Thermo Fisher Scientific); APC conjugated with Annexin V (Invitrogen™, Thermo Fisher Scientific); dimethylsulfoxide (Dinâmica, Sigma-Aldrich®); Dulbecco’s modified Eagle’s medium Ham F-12 (Merk, Sigma-Aldrich®); Dulbecco’s phosphate buffered saline (LGC Biotecnologia®); fetal bovine serum (Nova Biotecnologia®); gentamicin (Gibco™, Thermo Fisher Scientific); Hoechst (Invitrogen™, Thermo Fisher Scientific); propidium iodide (Invitrogen™, Thermo Fisher Scientific); Trypsin-EDTA solution (Gibco™, Thermo Fisher Scientific); 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide - MTT (Invitrogen™, Thermo Fisher Scientific).

Venom and fraction preparations

Crotalus durissus terrificus venom (CdtV) was obtained from the Serpentarium of the University of São Paulo, Ribeirão Preto School of Medicine, Ribeirão Preto, São Paulo, Brazil. The licenses related to access to Brazilian genetic resources for scientific purposes are the authorizations: CGEN/CNPq 010627/2011-1; IBAMA 27131-2 and CEBio UNIR-FIOCRUZ-RO (register CGEN A4D12CB and IBAMA/SISBIO 64385-1).

A total of 250 mg of the dry venom was dispersed in 2.5 mL of 0.05 M ammonium formate buffer (NH4HCO2) pH 3.5 and centrifuged at 755 xg for 10 min at room temperature (25 °C). The clear supernatant was then applied to a Sephadex G-75 column equilibrated with the same buffer; fractions of 0.1 mL/tube were collected and monitored at Abs 280 nm using an automatic fraction collector. Purification of crotoxin and subunits [crotoxin A (crotapotin) and crotoxin B (CB or Asp 49-PLA2)] was performed as described elsewhere with minimal changes [3030. Bercovici D, Chudziniski AM, Dias NO, Esteves MI, Hiraichi E, Oishi NY, Picarelli ZP, Rocha MC, Ueda CMPM, Yamanouye N, Raw I. Crotalus durissus terrificus venom. Mem Inst Butantan. 1987;49:69-78., 3131. de Oliveira DG, Toyama MH, Martins AM, Havt A, Nobre AC, Marangoni S, Câmara PR, Antunes E, de Nucci G, Beliam LO, Fonteles MC, Monteiro HS. Structural and biological characterization of a crotapotin isoform isolated from Crotalus durissus cascavella venom. Toxicon. 2003 Jul;42(1):53-62., 3232. Marchi-Salvador DP, Corrêa LC, Magro AJ, Oliveira CZ, Soares AM, Fontes MR. Insights into the role of oligomeric state on the biological activities of crotoxin: crystal structure of a tetrameric phospholipase A2 formed by two isoforms of crotoxin B from Crotalus durissus terrificus venom. Proteins. 2008 Aug 15;72(3):883-91., 3333. Carvalho LH, Teixeira LF, Zaqueo KD, Bastos JF, Nery NM, Setúbal SS, Pontes AS, Butzke D, Cavalcante W, Gallacci M, Fernandes CFC, Stabeli RG, Soares AM, Zuliani JP. Local and systemic effects caused by Crotalus durissus terrificus, Crotalus durissus collilineatus, and Crotalus durissus cascavella snake venoms in swiss mice. Rev Soc Bras Med Trop. 2019 Sep 5;52:e20180526. , 3434. Vieira LF, Magro AJ, Fernandes CA, de Souza BM, Cavalcante WL, Palma MS, Rosa JC, Fuly AL, Fontes MR, Gallacci M, Butzke DS, Calderon LA, Stábeli RG, Giglio JR, Soares AM. Biochemical, functional, structural and phylogenetic studies on Intercro, a new isoform phospholipase A2 from Crotalus durissus terrificus snake venom. Biochimie. 2013 Dec;95(12):2365-75., 3535. Sousa IDL, Barbosa AR, Salvador GHM, Frihling BEF, Santa-Rita PH, Soares AM, Pessôa HLF, Marchi-Salvador DP. Secondary hemostasis studies of crude venom and isolated proteins from the snake Crotalus durissus terrificus. Int J Biol Macromol. 2019 Jun 15;131:127-33. ]. The elution was monitored at an absorbance of 280 nm, manually collected, lyophilized, and stored at -20 °C.

Cell lines and cell culture

Mammary tumor samples were collected at the Veterinary Hospital of the School of Veterinary Medicine and Animal Science of UNESP, Botucatu. The fragments were placed in a culture medium (DMEM), added with 1% gentamicin and 0.5% antibiotic-antimycotic solution, and supplemented with 10% fetal bovine serum. For in vitro expansion, the collected neoplasm fragments measuring approximately one cm² were dissociated with the type IV collagenase enzyme for four hours. Plating was done at a concentration of 104 cells/mL in culture bottles with a volume equal to 25 mL. After achieving 80% confluence in the bottle, the cells were trypsinized and placed in new 75 cm² bottles with a filter. The cell lines used, UNESP-CM1 and UNESP-CM9, were previously established and characterized by Lainetti [3636. de Faria Lainetti P, Brandi A, Leis Filho AF, Prado MCM, Kobayashi PE, Laufer-Amorim R, Fonseca-Alves CE. Establishment and Characterization of Canine Mammary Gland Carcinoma Cell Lines With Vasculogenic Mimicry Ability in vitro and in vivo. Front Vet Sci. 2020 Oct 27;7:583874.]. UNESP-CM1 comes from a 12-year-old poodle and was classified as a solid carcinoma, grade II with tubular formation and HER2 overexpressing. UNESP-CM9 comes from a 12-year-old mixed breed and was classified as a tubulopapillary, grade II with tubular formation and HER2 overexpressing. The cells used were in their logarithmic growth phase in all experiments.

In vitro cytotoxicity assay

The colorimetric MTT assay was used to determine the cytotoxic effect of crotoxin, PLA2 (CB-PLA2), crotapotin, CdtV, and doxorubicin. For this, 1×104 cells/well of UNESP-CM1 and UNESP-CM9 were seeded in a 96-well plate at different concentrations of the compounds. After 24 hours of seeding in DMEM with 10% FBS, the media was replaced by DMEM without BFS plus the different concentrations of the tested compounds under the same conditions and for the same period (24 hours). For crotoxin, the doses tested were: 120, 240, 360, and 480 μg/mL. For CB-PLA2: 72.5, 145, 217.5, 290 μg/mL. For Crotapotin: 48, 96, 44, 192 μg/mL. For Crotalus durissus terrificus venom: 60, 120, 240, 480 μg/mL. For doxorubicin: 1.5, 3, 6, 12; 9, 18, 36, 72 μg/mL.

The MTT assay (Invitrogen™, Thermo Fisher Scientific, USA) was performed according to the manufacturer’s instructions, and Spectro colorimetric analysis was performed in a microplate reader (570 nm range). All the compounds were tested in four concentrations made in a serial dilution, guided by control groups, and executed in triplicate. GraphPad Prism 8.0.1 software was used to normalize the Spectro colorimetric data, plot a nonlinear regression, and determine the IC50 by the dose-response curve. The treated groups and control were compared by individual t-tests considering p < 0.05 to be significant.

Cellular migration

The inhibition potential for cellular migration was tested by a wound healing test after treatment with the IC50 doses of crotoxin, PLA2 (CB-PLA2), crotapotin, and doxorubicin. For this, the cells were seeded in a 6-well plate (4.5×105 cells/well) until they reached 80% confluence. After that, a 100 µL pipette tip was used to trace a linear wound. The plates were double-washed with DPBS (500 µL) and 5 min agitation to remove the detached cells.

For reference, each well was photographed at the time the wound was made and after 24 hours. During this time, the cells were incubated in fresh DMEM (without FBS), and the compound testing dosage was added. Wound healing was measured in five different regions using the GIMP 2.10.14 program. The mean distance for each cell line was calculated as the mean and standard deviation. The treated groups and control were compared by individual T-tests considering p < 0,05.

Apoptosis analysis

The cell death analysis of UNESP-CM1 and UNESP-CM9 was performed after 24 hours of treatment with the IC50 dose of all the tested compounds. The samples were suspended in a medium containing calcium (buffer solution) for analysis of apoptosis. To this end, 5 µL of APC-conjugated annexin V (Becton Dickinson and Company), 5 µL (1.5 µM final concentration) of propidium iodide (Becton Dickinson and Company), and 5 µL (7 µM final concentration) of Hoechst (Sigma, USA) were added to the cell suspensions.

All samples were incubated in the dark for 15 min at room temperature, and flow cytometry assessment was performed with a final concentration of 1×106 cells/mL in Fortessa LSR equipment (Becton Dickinson, Mountain View, CA, USA). The filter configurations for the PMTs measuring fluorescence emission of the applied fluorochromes were 694/50 nm (IP), 660/20 nm (Annexin-APC), and 450/50 nm (Hoechst 33342). The acquisition rate was 800 events per second, and at least 1×104 cells were analyzed per sample.

Data were generated in a contour plot graph including axis < 0t (biexponential), making all events visible and properly compensated through BD FACSDiva TM software v6.1 (Becton Dickinson).

Cytotoxicity analysis of crotoxin in normal mammary gland cells

Normal canine mammary glands were collected for histopathological examination and cell culture, and the fragment was processed by enzymatic dissociation with collagenase type IV for 4 hours. The resultant material was filtered and centrifuged in a controlled environment, with subsequent seeding in a 25 cm2 cell culture flask with DMEM until 80% confluence was obtained. Cytotoxicity analysis was tested for crotoxin in the same manner as for the tumor cell lines.

Data analysis

The IC50 values of all the tested compounds were obtained by plotting the spectrum colorimetric data in a dose-response curve after the application of data normalization and a nonlinear regression method in GraphPad Prism 8.0.1 software. The values presented are the mean and standard deviation of triplicate tests, and statistical significance (p < 0,05) was obtained by a comparison of each tested group with the control (vehicle) by independent t-tests and ANOVA.

For the cell migration analysis, the mean and standard deviation were calculated. The wound healing (difference) was taken by the formula D0-D1, of which D0 was the first measure, and D1 was the measure compared to the initial moment. The treated groups and control were compared using the Mann‒Whitney test, considering p < 0.05 as an indicator of statistical significance, and this was done in GraphPad Prism 8.0.1 software.

Results

Venom and fraction preparations

Figure 1 shows the chromatographic profile of the venom of Crotalus d. terrificus (Figure 1A ), with five fractions, F4 being the crotoxin, as shown in the electrophoresis gel (Figure 1B ).

Figure 1.
Chromatographic profile of the Crotalus durissus terrificus venom pool, where four fractions were observed, the third peak was suggestive of crotoxin. On the X axis, the time measured in minutes, on the Y axis on the right, the concentration of eluent B and on the Y axis on the left, the optical density expressed in AU (280 nm). On the left, the SDS-PAGE gel of the third peak, with the presence of two distinct bands, suggestive of crotapotin (8.9 kDa) and PLA2 (14.3 kDa).

Cellular Viability after toxin exposure

For both canine mammary carcinoma cell lines (UNESP-CM1 and UNESP-CM9), there was a decline in cellular metabolic activity by the increase in CTX, CB-PLA2, crotalic venom, and doxorubicin in a dose-dependent manner (Table 1 and Figure 2). For the acid component of crotoxin (crotapotin), we did not find an antitumoral effect.

Table 1.
IC50 of the drugs tested in UNESP-CM1 and UNESP-CM9, two canine mammary carcinoma cell lines.

Figure 2.
Cytotoxicity assay (MTT - 24 h) on the canine mammary carcinoma cell lines UNESP-CM1 (left) and UNESP-CM9 (right). ( A) Note the decrease in cell viability as there is an increase in crotoxin (CTX) concentrations for both cell lines. ( B) Note the decrease in cell viability as there is an increase in CB-PLA2 concentrations for both cell lines. ( C) Note the absence of a toxic effect of crotapotin on both cell lines. ( D) Note the decrease in cell viability as there is an increase in the concentrations of Crotalus durissus terrificus venom in both cell lines. ( E) Note the decrease in cell viability with doxorubicin in a dose-dependent manner in both canine mammary carcinoma cell lines.

Cellular migration

For UNESP-CM1 and UNESP-CM9, when comparing the mean of closure of the wound difference between 0 h and 24 h, Crotoxin, CB-PLA2, and doxorubicin showed an effect on delaying wound healing, but with no significant difference ( Figures 3 and 4 and Table 2). Crotapotin did not inhibit cell migration.

Figure 3.
Wound healing test in the tested and control groups at 0 and 24 h in the UNESP-CM1 canine mammary gland carcinoma cell line.

Figure 4.
Wound healing test in the tested and control groups at 0 and 24 h in the UNESP-CM9 canine mammary gland carcinoma cell line.

Table 2.
Wound measurement and healing percentage in 24 h intervals.

Mechanism of cell death

Apoptosis was the main cell death mechanism for crotoxin and doxorubicin. The compound with the highest apoptotic potential was crotoxin. Late apoptosis was more frequently detected than premature apoptosis in all samples except CB-PLA2 for UNESP-CM9 (Figure 5).

Figure 5.
Apoptosis Assay (Annexin V x Propidium Iodide): Note that crotoxin and crotapotin induced apoptosis of the UNESP-CM1 cell line. CB-PLA2 preferentially induced death by necrosis. Note that crotoxin and CB-PLA2 induced apoptosis of the UNESP-CM9 cell line. Crotapotin preferentially induced death by necrosis.

Normal breast cell cytotoxicity analysis

For normal canine mammary cells, 312 µg/mL crotoxin did not show a cytotoxic effect when compared to the same cells treated with DPBS (vehicle) or DMEM (Figure 6). When a similar dose was used in canine mammary tumor cells (UNESP CM-1 and UNESP CM-9), cytotoxic effects and more than 50% mortality were observed in these cells.

Figure 6.
Crotoxin effect on normal canine mammary cells at a dose of 312 µg/mL.

Discussion

Crotalus durissus terrificus venom has been tested in vitro as an antitumoral drug in breast cancer cell lines [1818. Almeida CF, Amaral C, Augusto TV, Correia-da-Silva G, Marques de Andrade C, Torqueti MR, Teixeira N. The anti-cancer potential of crotoxin in estrogen receptor-positive breast cancer: Its effects and mechanism of action. Toxicon. 2021 Sep;200:69-77., 3737. Muller SP, Silva VAO, Silvestrini AVP, de Macedo LH, Caetano GF, Reis RM, Mazzi MV. Crotoxin from Crotalus durissus terrificus venom: In vitro cytotoxic activity of a heterodimeric phospholipase A2on human cancer-derived cell lines. Toxicon. 2018 Dec 15;156:13-22.] but not yet in canine mammary carcinoma cells. Considering that canine mammary tumors are a good natural model for comparative oncology studies, we tested Crotalus durissus terrificus venom in two canine mammary carcinoma cell lines, with results similar to human breast cancer cell lines. Other human cancer cell lines have been tested for the antitumoral effects of crotoxin, such as lung, pancreas, cervix, colon, kidney, ovary, esophagus, brain, melanoma, glioma, and squamous cell carcinoma [3838. Chaisakul J, Hodgson WC, Kuruppu S, Prasongsook N. Effects of Animal Venoms and Toxins on Hallmarks of Cancer. J Cancer. 2016Jul 15;7(11):1571-8., 3939. Han R, Liang H, Qin Zh, Liu CY. Crotoxin induces apoptosis and autophagy in human lung carcinoma cells in vitro via activation of the p38MAPK signaling pathway. Acta Pharmacol Sin. 2014 Oct;35(10):1323-32., 4040. He Jk, Wu Xs, Wang Y, Han R, Qin ZH, Xie Y. Growth inhibitory effects and molecular mechanisms of crotoxin treatment in esophageal Eca-109 cells and transplanted tumors in nude mice. Acta Pharmacol Sin. 2013 Feb;34(2):295-300. , 4141. Rodrigues RS, Izidoro LF, de Oliveira RJ Jr, Sampaio SV, Soares AM, Rodrigues VM. Snake venom phospholipases A2: a new class of antitumor agents. Protein Pept Lett. 2009;16(8):894-8., 4242. da Rocha RG, Santos EMS, Tanaka-Azevedo AM, Serino-Silva C, Souza MG, Gomes ESB, Guimarães FAD, Silveira LH, Santos SHS, de Paula AMB, Gomez RS, Guimarães ALS, Farias LC. The antineoplastic potential of crotoxin isolated from Crotalus durissus terrificus snake venom on oral squamous cell carcinoma. Toxicon. 2023 Jan 1;221:106965.].

Crotoxin is the main toxin present in rattlesnake venom and is composed of two subunits: an acidic subunit (crotapotin) and one basic subunit (CB-PLA2). In our study, the basic subunit was responsible for the cytotoxic activity of crotoxin, showing an IC50 of 25.43 and 57.98 μg/mL. In contrast, the acidic subunit did not show such effects at a 192 μg/mL dose. This result is in accordance with the study carried out by Corin [4343. Corin RE, Viskatis LJ, Vidal JC, Etcheverry MA. Cytotoxicity of crotoxin on murine erythroleukemia cells in vitro. Invest New Drugs. 1993 Feb;11(1):11-5.] with MEL cells from murine erythroleukemia, where crotoxin showed an IC50 of 0.8-1.0 µg/mL for these cells, without showing toxicity of crotapotin at 20 µg/mL. In addition, the IC50 of isolated CB-PLA2 is lower than that of crotoxin, which allowed us to perceive that this secluded molecule may be more adequate because it has a minor molecular weight, and this fact directly influences the use of drug transporters and penetration into cell membranes, optimizing their effectiveness as a therapeutic agent.

The effects of crotoxin in normal canine breast cells at a 312 µg/mL dose showed no toxicity for these cells, as proven by other authors. Almeida [1818. Almeida CF, Amaral C, Augusto TV, Correia-da-Silva G, Marques de Andrade C, Torqueti MR, Teixeira N. The anti-cancer potential of crotoxin in estrogen receptor-positive breast cancer: Its effects and mechanism of action. Toxicon. 2021 Sep;200:69-77.] showed that crotoxin is a secure compound for non-tumor cells (HFF-1) at 100 μg/mL doses. Rudd [4444. Rudd CJ, Viskatis LJ, Vidal JC, Etcheverry MA. In vitro comparison of cytotoxic effects of crotoxin against three human tumors and a normal human epidermal keratinocyte cell line. Invest New Drugs. 1994;12(3):183-4.] demonstrated low toxicity of CTX in normal cells, indicating selectivity for tumor cells. In this experiment with normal canine mammary cells, we only use crotoxin, and this is justified since the subunits are mixed and present in the complete protein, adding to the already known toxic effects of doxorubicin in normal cells.

Muller [3737. Muller SP, Silva VAO, Silvestrini AVP, de Macedo LH, Caetano GF, Reis RM, Mazzi MV. Crotoxin from Crotalus durissus terrificus venom: In vitro cytotoxic activity of a heterodimeric phospholipase A2on human cancer-derived cell lines. Toxicon. 2018 Dec 15;156:13-22.] also showed this specific cytotoxicity using mouse fibroblasts (3T3) and human keratinocytes (HaCaT). At a 30 μg/mL dose, crotoxin was not toxic to these cells, which are strongly affected by antineoplastic chemotherapies [4545. Plescia J, Salz W, Xia F, Pennati M, Zaffaroni N, Daidone MG, Meli M, Dohi T, Fortugno P, Nefedova Y, Gabrilovich DI, Colombo G, Altieri DC. Rational design of shepherdin, a novel anticancer agent. Cancer Cell. 2005 May;7(5):457-68.]. However, it showed a significant drop in viability in pancreatic cancer (PSN-1 and PANC-1), esophageal cancer (Kyse 30), cervical cancer (HeLa), and glioma (GAMG, HCB 151 and U373) cell lines. The most resistant cell lines were cervical (SiHa) and esophageal (KYSE270) carcinoma. Although crotoxin is toxic to all cell lines, it presents a very heterogeneous response, even in the same type of tumor, which corroborates our study where the IC50 values for two different canine mammary tumor cell lines were 172.08 µg/mL and 310.80 µg/mL for UNESP-CM1 and UNESP-CM9, respectively.

In our study, crotoxin inhibited cell migration, presenting a lower healing percentage than the control group for CM1 8.67% and CM9 5.58%, compared to 13.20% and 15.24% in the control group for the respective cell lines. In the same test, Da Rocha [4242. da Rocha RG, Santos EMS, Tanaka-Azevedo AM, Serino-Silva C, Souza MG, Gomes ESB, Guimarães FAD, Silveira LH, Santos SHS, de Paula AMB, Gomez RS, Guimarães ALS, Farias LC. The antineoplastic potential of crotoxin isolated from Crotalus durissus terrificus snake venom on oral squamous cell carcinoma. Toxicon. 2023 Jan 1;221:106965.] also demonstrated inhibition of migration in oral squamous carcinoma cells, making it a promising compound in the oncology field since migration is one of the bases of the mechanism of metastasis.

The crotoxin in this experiment showed that apoptosis was the most prevalent cell death mechanism. We observed 56% apoptosis versus 22.9% necrosis for UNESP-CM1 and 35.2% apoptosis versus 24.5% necrosis for UNESP-CM9 cells. A similar method has already been identified in other models of cancer cells by other authors. Han et al. (201439. Han R, Liang H, Qin Zh, Liu CY. Crotoxin induces apoptosis and autophagy in human lung carcinoma cells in vitro via activation of the p38MAPK signaling pathway. Acta Pharmacol Sin. 2014 Oct;35(10):1323-32.) in human lung squamous carcinoma (SK-MES-1) and Ye [4646. Ye B, Xie Y, Qin ZH, Wu JC, Han R, He JK. Anti-tumor activity of CrTX in human lung adenocarcinoma cell line A549. Acta Pharmacol Sin. 2011 Nov;32(11):1397-401.] in human lung adenocarcinoma (A549) demonstrated apoptosis by increasing p38 MAPK and caspase 3. Another mechanism of action of crotoxin was the reduction of angiogenesis and tumor growth in a xenograft model [4646. Ye B, Xie Y, Qin ZH, Wu JC, Han R, He JK. Anti-tumor activity of CrTX in human lung adenocarcinoma cell line A549. Acta Pharmacol Sin. 2011 Nov;32(11):1397-401.]. He [ 4040. He Jk, Wu Xs, Wang Y, Han R, Qin ZH, Xie Y. Growth inhibitory effects and molecular mechanisms of crotoxin treatment in esophageal Eca-109 cells and transplanted tumors in nude mice. Acta Pharmacol Sin. 2013 Feb;34(2):295-300. ] showed that CTX induces caspase 3 in human esophageal carcinoma (Eca-109). Almeida [1818. Almeida CF, Amaral C, Augusto TV, Correia-da-Silva G, Marques de Andrade C, Torqueti MR, Teixeira N. The anti-cancer potential of crotoxin in estrogen receptor-positive breast cancer: Its effects and mechanism of action. Toxicon. 2021 Sep;200:69-77.] demonstrated in ER+ breast cancer cells that CTX induces apoptosis mediated by caspase-8. Da Rocha [4242. da Rocha RG, Santos EMS, Tanaka-Azevedo AM, Serino-Silva C, Souza MG, Gomes ESB, Guimarães FAD, Silveira LH, Santos SHS, de Paula AMB, Gomez RS, Guimarães ALS, Farias LC. The antineoplastic potential of crotoxin isolated from Crotalus durissus terrificus snake venom on oral squamous cell carcinoma. Toxicon. 2023 Jan 1;221:106965.] showed reduced cell viability by increasing DNA damage, in addition to reducing the expression of MMP9, MM2, and COL1A1 (proteins related to invasion and metastasis).

Similar to other medicines earned by snake venom sources, the multimodal benefits promised exerted by crotoxin in canine oncologic patient treatment bring new possibilities. The results of a phase I clinical trial of CTX performed in patients with untreatable tumors were published in 2002. Limited neurological toxic effects were observed, but all of these effects disappeared completely during the study. Of the 23 patients, two had a tumor mass reduction greater than 50%, one with a measurable tumor in the vaginal region, had a reduced tumor mass allowing for surgical removal, and one patient had a complete response [2727. Cura JE, Blanzaco DP, Brisson C, Cura MA, Cabrol R, Larrateguy L, Mendez C, Sechi JC, Silveira JS, Theiller E, de Roodt AR, Vidal JC. Phase I and pharmacokinetics study of crotoxin (cytotoxic PLA(2), NSC-624244) in patients with advanced cancer. Clin Cancer Res. 2002 Apr;8(4):1033-41.].

Conclusion

We confirmed that crotoxin can exert cytotoxic effects in canine mammary carcinoma cells (UNESP-CM1 and UNESP-CM9) in addition to being noncytotoxic to normal canine breast cells. This use of crotoxin in canine mammary carcinoma cell lines reveals the success of this toxin in promoting apoptosis, opening a promising opportunity for further studies in vivo, using the dog as a natural model for mammary carcinoma clinical trials.

Abbreviations

CTX: crotoxin; CB-PLA2: PLA2 of crotoxin; CdtV: Crotalus durissus terrificus venom; DMSO: dimethylsulfoxide; DMEM: Dulbecco’s modified Eagle’s medium; DPBS: Dulbecco’s phosphate buffered saline; FBS: fetal bovine serum.

Acknowledgments

The authors express their gratitude to Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES), Fundação de Amparo à Pesquisa do Estado de Rondônia (FAPERO), Instituto Federal de Rondônia (IFRO) and Fundação Oswaldo Cruz (FIOCRUZ)/FIOTEC. The authors thank the Network Technological Platforms from FIOCRUZ (https://plataformas.fiocruz.br/) for the support and financing of the services provided by the Toxinological Bioprospecting and Molecular Interaction facilities/Fiocruz Rondonia and Programa de Excelência em Pesquisa da Fiocruz Rondonia - PROEP.

References

  • 1. Nakagaki KY, Nunes MM, Garcia APV, Nunes FC, Schmitt F, Cassali GD. Solid Carcinoma of the Canine Mammary Gland: a Histological Type or Tumour Cell Arrangement? J Comp Pathol. 2022 Jan;190:1-12.
  • 2. Nunes FC, Campos CB, Teixeira SV, Bertagnolli AC, Lavalle GE, Cassali GD. Epidemiological, clinical and pathological evaluation of overall survival in canines with mammary neoplasms. Arq Bras Med Vet Zootec. 2018 Nov-Dez;70(6):1714-22.
  • 3. Instituto Nacional de Cancer José Alencar Gomes Silva. Estimativa 2023: incidência do Câncer no Brasil. Rio de Janeiro: INCA, 2022. Disponível em: https://www.gov.br/inca/ptbr/assuntos/cancer/numeros/estimativa.
    » https://www.gov.br/inca/ptbr/assuntos/cancer/numeros/estimativa.
  • 4. Kwon JY, Moskwa N, Kang W, Fan TM, Lee C. Canine as a Comparative and Translational Model for Human Mammary Tumor. J Breast Cancer. 2023 Feb;26(1):1-13.
  • 5. Raffo-Romero A, Aboulouard S, Bouchaert E, Rybicka A, Tierny D, Hajjaji N, Fournier I, Salzet M, Duhamel M. Establishment and characterization of canine mammary tumoroids for translational research. BMC Biol. 2023 Feb 3;21(1):23.
  • 6. Antuofermo E, Miller MA, Pirino S, Xie J, Badve S, Mohammed SI. Spontaneous mammary intraepithelial lesions in dogs--a model of breast cancer. Cancer Epidemiol Biomarkers Prev. 2007 Nov;16(11):2247-56.
  • 7. Huang M, Lu JJ, Ding J. Natural Products in Cancer Therapy: Past, Present and Future. Nat Prod Bioprospect. 2021;11:5-13.
  • 8. Newman DJ, Cragg GM. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019. J Nat Prod. 2020 Mar 27;83(3):770-803.
  • 9. Alves BFA, Ferreira RS Jr. Antineoplastic properties and pharmacological applications of Crotalus durissus terrificus snake venom. Rev Soc Bras Med Trop. 2022 Dec 16;55:e0323-2022.
  • 10. Abbade LPF, Barraviera SRCS, Silvares MRC, Lima ABBCO, Haddad GR, Gatti MAN, Medolago NB, Rigotto Carneiro MT, Dos Santos LD, Ferreira RS Jr, Barraviera B. Treatment of Chronic Venous Ulcers With Heterologous Fibrin Sealant: A Phase I/II Clinical Trial. Front Immunol. 2021 Feb 23;12:627541.
  • 11. Diniz-Sousa R, Caldeira CADS, Pereira SS, Da Silva SL, Fernandes PA, Teixeira LMC, Zuliani JP, Soares AM. Therapeutic applications of snake venoms: An invaluable potential of new drug candidates. Int J Biol Macromol. 2023 May 31;238:124357.
  • 12. Mohamed Abd El-Aziz T, Garcia Soares A, Stockand JD. Snake Venoms in Drug Discovery: Valuable Therapeutic Tools for Life Saving. Toxins (Basel). 2019 Sep 25;11(10):564.
  • 13. Oliveira AL, Viegas MF, da Silva SL, Soares AM, Ramos MJ, Fernandes PA. The chemistry of snake venom and its medicinal potential. Nat Rev Chem. 2022 Jun 10;6:451-69.
  • 14. Shahbazi B, Najafabadi ZS, Goudarzi H, Sajadi M, Tahoori F, Bagheri M. Cytotoxic effects of Pseudocerastes persicus venom and its HPLC fractions on lung cancer cells. J Venom Anim Toxins incl Trop Dis. 2019 Sep 16;25:e20190009. doi: 10.1590/1678-9199-JVATITD-2019-0009. PMID: 31555336; PMCID: PMC6748451.
    » https://doi.org/10.1590/1678-9199-JVATITD-2019-0009
  • 15. Calderon LA, Sobrinho JC, Zaqueo KD, de Moura AA, Grabner AN, Mazzi MV, Marcussi S, Nomizo A, Fernandes CF, Zuliani JP, Carvalho BM, da Silva SL, Stábeli RG, Soares AM. Antitumoral activity of snake venom proteins: new trends in cancer therapy. Biomed Res Int. 2014;2014:203639.
  • 16. Macedo JM, Lima AM, Kayano AM, Souza MF, Oliveira IS, Garay AFG, Rocha AM, Zuliani JP, Soares AM. Literature review on Crotalus durissus terrificus toxins: From a perspective of structural biology and therapeutic applications. Curr Protein Pept Sci. 2023;24(7):536-50.
  • 17. Sartim MA, Menaldo DL, Sampaio SV. Immunotherapeutic potential of Crotoxin: anti-inflammatory and immunosuppressive properties. J Venom Anim Toxins incl Trop Dis. 2018 Dec 17;24:39. doi: 10.1186/s40409-018-0178-3. PMID: 30564276; PMCID: PMC6296157.
    » https://doi.org/10.1186/s40409-018-0178-3
  • 18. Almeida CF, Amaral C, Augusto TV, Correia-da-Silva G, Marques de Andrade C, Torqueti MR, Teixeira N. The anti-cancer potential of crotoxin in estrogen receptor-positive breast cancer: Its effects and mechanism of action. Toxicon. 2021 Sep;200:69-77.
  • 19. de Oliveira LA, Ferreira RS Jr, Barraviera B, de Carvalho FCT, de Barros LC, Dos Santos LD, Pimenta DC. Crotalus durissus terrificus crotapotin naturally displays preferred positions for amino acid substitutions. J Venom Anim Toxins incl Trop Dis. 2017 Nov 28;23:46. doi: 10.1186/s40409-017-0136-5. PMID: 29209361; PMCID: PMC5704381.
    » https://doi.org/10.1186/s40409-017-0136-5
  • 20. Sampaio SC, Hyslop S, Fontes MR, Prado-Franceschi J, Zambelli VO, Magro AJ, Brigatte P, Gutierrez VP, Cury Y. Crotoxin: novel activities for a classic beta-neurotoxin. Toxicon. 2010 Jun 1;55(6):1045-60.
  • 21. Fortes-Dias CL, Lin Y, Ewell J, Diniz CR, Liu TY. A phospholipase A2 inhibitor from the plasma of the South American rattlesnake ( Crotalus durissus terrificus). Protein structure, genomic structure, and mechanism of action. J Biol Chem. 1994 Jun 3;269(22):15646-51.
  • 22. Sant’Anna MB, Lopes FSR, Kimura LF, Giardini AC, Sant’Anna OA, Picolo G. Crotoxin Conjugated to SBA-15 Nanostructured Mesoporous Silica Induces Long-Last Analgesic Effect in the Neuropathic Pain Model in Mice. Toxins (Basel). 2019 Nov 20;11(12):679.
  • 23. Teixeira NB, Sant’Anna MB, Giardini AC, Araujo LP, Fonseca LA, Basso AS, Cury Y, Picolo G. Crotoxin down-modulates pro-inflammatory cells and alleviates pain on the MOG35-55-induced experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis. Brain Behav Immun. 2020 Feb;84:253-68.
  • 24. Sartim MA, Menaldo DL, Sampaio SV. Immunotherapeutic potential of Crotoxin: anti-inflammatory and immunosuppressive properties. J Venom Anim Toxins incl Trop Dis. 2018;24:39. https://doi.org/10.1186/s40409-018-0178-3
    » https://doi.org/10.1186/s40409-018-0178-3
  • 25. Wolz-Richter S, Esser KH, Hess A. Antinociceptive activity of crotoxin in the central nervous system: a functional Magnetic Resonance Imaging study. Toxicon. 2013 Nov;74:44-55.
  • 26. Almeida TC, Ribeiro Silva LM, Boaventura de Oliveira AM, Lopes FSR, Sant’Anna MB, Picolo G. Cytotoxic effect of crotoxin on cancer cells and its antitumoral effects correlated to tumor microenvironment: A review. Int J Biol Macromol. 2023 May 15;242(Pt 2):124892.
  • 27. Cura JE, Blanzaco DP, Brisson C, Cura MA, Cabrol R, Larrateguy L, Mendez C, Sechi JC, Silveira JS, Theiller E, de Roodt AR, Vidal JC. Phase I and pharmacokinetics study of crotoxin (cytotoxic PLA(2), NSC-624244) in patients with advanced cancer. Clin Cancer Res. 2002 Apr;8(4):1033-41.
  • 28. Medioni J, Brizard M, Elaidi R, Reid PF, Benlhassan K, Bray D. Innovative design for a phase 1 trial with intra-patient dose escalation: The Crotoxin study. Contemp Clin Trials Commun. 2017 Jul 23;7:186-8.
  • 29. Wang J, Qin X, Zhang Z, Chen M, Wang Y, Gao B. Crotoxin suppresses the tumorigenic properties and enhances the antitumor activity of Iressa® (gefinitib) in human lung adenocarcinoma SPCA-1 cells. Mol Med Rep. 2014 Dec;10(6):3009-14.
  • 30. Bercovici D, Chudziniski AM, Dias NO, Esteves MI, Hiraichi E, Oishi NY, Picarelli ZP, Rocha MC, Ueda CMPM, Yamanouye N, Raw I. Crotalus durissus terrificus venom. Mem Inst Butantan. 1987;49:69-78.
  • 31. de Oliveira DG, Toyama MH, Martins AM, Havt A, Nobre AC, Marangoni S, Câmara PR, Antunes E, de Nucci G, Beliam LO, Fonteles MC, Monteiro HS. Structural and biological characterization of a crotapotin isoform isolated from Crotalus durissus cascavella venom. Toxicon. 2003 Jul;42(1):53-62.
  • 32. Marchi-Salvador DP, Corrêa LC, Magro AJ, Oliveira CZ, Soares AM, Fontes MR. Insights into the role of oligomeric state on the biological activities of crotoxin: crystal structure of a tetrameric phospholipase A2 formed by two isoforms of crotoxin B from Crotalus durissus terrificus venom. Proteins. 2008 Aug 15;72(3):883-91.
  • 33. Carvalho LH, Teixeira LF, Zaqueo KD, Bastos JF, Nery NM, Setúbal SS, Pontes AS, Butzke D, Cavalcante W, Gallacci M, Fernandes CFC, Stabeli RG, Soares AM, Zuliani JP. Local and systemic effects caused by Crotalus durissus terrificus, Crotalus durissus collilineatus, and Crotalus durissus cascavella snake venoms in swiss mice. Rev Soc Bras Med Trop. 2019 Sep 5;52:e20180526.
  • 34. Vieira LF, Magro AJ, Fernandes CA, de Souza BM, Cavalcante WL, Palma MS, Rosa JC, Fuly AL, Fontes MR, Gallacci M, Butzke DS, Calderon LA, Stábeli RG, Giglio JR, Soares AM. Biochemical, functional, structural and phylogenetic studies on Intercro, a new isoform phospholipase A2 from Crotalus durissus terrificus snake venom. Biochimie. 2013 Dec;95(12):2365-75.
  • 35. Sousa IDL, Barbosa AR, Salvador GHM, Frihling BEF, Santa-Rita PH, Soares AM, Pessôa HLF, Marchi-Salvador DP. Secondary hemostasis studies of crude venom and isolated proteins from the snake Crotalus durissus terrificus Int J Biol Macromol. 2019 Jun 15;131:127-33.
  • 36. de Faria Lainetti P, Brandi A, Leis Filho AF, Prado MCM, Kobayashi PE, Laufer-Amorim R, Fonseca-Alves CE. Establishment and Characterization of Canine Mammary Gland Carcinoma Cell Lines With Vasculogenic Mimicry Ability in vitro and in vivo Front Vet Sci. 2020 Oct 27;7:583874.
  • 37. Muller SP, Silva VAO, Silvestrini AVP, de Macedo LH, Caetano GF, Reis RM, Mazzi MV. Crotoxin from Crotalus durissus terrificus venom: In vitro cytotoxic activity of a heterodimeric phospholipase A2on human cancer-derived cell lines. Toxicon. 2018 Dec 15;156:13-22.
  • 38. Chaisakul J, Hodgson WC, Kuruppu S, Prasongsook N. Effects of Animal Venoms and Toxins on Hallmarks of Cancer. J Cancer. 2016Jul 15;7(11):1571-8.
  • 39. Han R, Liang H, Qin Zh, Liu CY. Crotoxin induces apoptosis and autophagy in human lung carcinoma cells in vitro via activation of the p38MAPK signaling pathway. Acta Pharmacol Sin. 2014 Oct;35(10):1323-32.
  • 40. He Jk, Wu Xs, Wang Y, Han R, Qin ZH, Xie Y. Growth inhibitory effects and molecular mechanisms of crotoxin treatment in esophageal Eca-109 cells and transplanted tumors in nude mice. Acta Pharmacol Sin. 2013 Feb;34(2):295-300.
  • 41. Rodrigues RS, Izidoro LF, de Oliveira RJ Jr, Sampaio SV, Soares AM, Rodrigues VM. Snake venom phospholipases A2: a new class of antitumor agents. Protein Pept Lett. 2009;16(8):894-8.
  • 42. da Rocha RG, Santos EMS, Tanaka-Azevedo AM, Serino-Silva C, Souza MG, Gomes ESB, Guimarães FAD, Silveira LH, Santos SHS, de Paula AMB, Gomez RS, Guimarães ALS, Farias LC. The antineoplastic potential of crotoxin isolated from Crotalus durissus terrificus snake venom on oral squamous cell carcinoma. Toxicon. 2023 Jan 1;221:106965.
  • 43. Corin RE, Viskatis LJ, Vidal JC, Etcheverry MA. Cytotoxicity of crotoxin on murine erythroleukemia cells in vitro Invest New Drugs. 1993 Feb;11(1):11-5.
  • 44. Rudd CJ, Viskatis LJ, Vidal JC, Etcheverry MA. In vitro comparison of cytotoxic effects of crotoxin against three human tumors and a normal human epidermal keratinocyte cell line. Invest New Drugs. 1994;12(3):183-4.
  • 45. Plescia J, Salz W, Xia F, Pennati M, Zaffaroni N, Daidone MG, Meli M, Dohi T, Fortugno P, Nefedova Y, Gabrilovich DI, Colombo G, Altieri DC. Rational design of shepherdin, a novel anticancer agent. Cancer Cell. 2005 May;7(5):457-68.
  • 46. Ye B, Xie Y, Qin ZH, Wu JC, Han R, He JK. Anti-tumor activity of CrTX in human lung adenocarcinoma cell line A549. Acta Pharmacol Sin. 2011 Nov;32(11):1397-401.
  • Availability of data and materials

    The datasets generated during and/or analyzed during the current study are available from the corresponding author upon reasonable request.
  • Funding

    This work was supported by grant 2020/15528-4, São Paulo Research Foundation (FAPESP).
  • Ethical approval

    Approved by CEUA FMVZ UNESP Botucatu 0470/2023.
  • Consent for publication

    Not applicable.

Publication Dates

  • Publication in this collection
    18 Mar 2024
  • Date of issue
    2024

History

  • Received
    24 Aug 2023
  • Accepted
    16 Feb 2024
Centro de Estudos de Venenos e Animais Peçonhentos (CEVAP/UNESP) Av. Universitária, 3780, Fazenda Lageado, Botucatu, SP, CEP 18610-034, Brasil, Tel.: +55 14 3880-7693 - Botucatu - SP - Brazil
E-mail: editorial.jvatitd@unesp.br