You are on page 1of 19

J O U R NA L OF PR O TE O MI CS 73 ( 20 1 0 ) 1 7 5 8– 1 7 7 6

available at www.sciencedirect.com

www.elsevier.com/locate/jprot

Snake venomics and antivenomics of Crotalus durissus


subspecies from Brazil: Assessment of geographic variation
and its implication on snakebite management

Johara Boldrini-Françaa , Carlos Corrêa-Nettob , Marliete M.S. Silvac , Renata S. Rodriguesd ,


Pilar De La Torree , Alicia Péreze , Andreimar M. Soaresa , Russolina B. Zingalib ,
Romildo A. Nogueirac , Veridiana M. Rodriguesd , Libia Sanze , Juan J. Calvetee,⁎
a
Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
b
Instituto de Bioquímica Médica, Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem-INBEB and Rede Proteomica do
Rio de Janeiro-Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
c
Departamento de Morfologia e Fisiologia Animal, Universidade Federal Rural de Pernambuco, Recife, Brazil
d
Instituto de Genética e Bioquímica, Universidade Federal de Uberlândia, Uberlândia, Brazil
e
Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (CSIC), Valencia, Spain

AR TIC LE I N FO ABS TR ACT

Article history: We report the comparative proteomic and antivenomic characterization of the venoms of
Received 10 May 2010 subspecies cascavella and collilineatus of the Brazilian tropical rattlesnake Crotalus durissus.
Accepted 3 June 2010 The venom proteomes of C. d. collilineatus and C. d. cascavella comprise proteins in the
range of 4–115 kDa belonging to 9 and 8 toxin families, respectively. Collilineatus and
cascavella venoms contain 20–25 main toxins belonging to the following protein families:
Keywords: disintegrin, PLA2, serine proteinase, cysteine-rich secretory protein (CRISP), vascular
Crotalus durissus subspecies endothelial growth factor-like (VEGF), L-amino acid oxidase, C-type lectin-like, and snake
Snake venom proteomics venom metalloproteinase (SVMP). As judged by reverse-phase HPLC and mass
Geographic venom variation spectrometry, cascavella and collilineatus share about 90% of their venom proteome.
Individual venom variability However, the relative occurrence of the toxin families departs among the two C. durissus
Crotamine subspecies venoms. The most notable difference is the presence of the myotoxin crotamine
Mass spectrometry in some C. d. collilineatus specimens (averaging 20.8% of the total proteins of pooled venom),
Antivenomics which is absent in the venom of C. d. cascavella. On the other hand, the neurotoxic PLA2
Anti-crotalic antivenom crotoxin represents the most abundant protein in both C. durissus venoms, comprising 67.4%
of the toxin proteome in C. d. collilineatus and 72.5% in C. d. cascavella. Myotoxic PLA2s are also
present in the two venoms albeit in different relative concentrations (18.1% in C. d. cascavella
vs. 4.6% in C. d. collilineatus). The venom composition accounts for the clinical manifestations
caused by C. durissus envenomations: systemic neurotoxicity and myalgic symptoms and
coagulation disturbances, frequently accompanied by myoglobinuria and acute renal
failure. The overall compositions of C. d. subspecies cascavella and collilineatus venoms
closely resemble that of C. d. terrificus, supporting the view that these taxa can be considered
geographical variations of the same species. Pooled venom from adult C.d. cascavella

⁎ Corresponding author. Instituto de Biomedicina de Valencia, CSIC, Jaume Roig 11, 46010 Valencia, Spain. Tel.: +34 96 339 1778; fax: +34 96
369 0800.
E-mail address: jcalvete@ibv.csic.es (J.J. Calvete).

1874-3919/$ – see front matter © 2010 Elsevier B.V. All rights reserved.
doi:10.1016/j.jprot.2010.06.001
J O U R NA L OF PR O TE O MI CS 7 3 ( 2 01 0 ) 1 7 5 8– 1 7 7 6 1759

and neonate C.d. terrificus lack crotamine, whereas this skeletal muscle cell membrane
depolarizing inducing myotoxin accounts for ∼20% of the total toxins of venom pooled
from C.d. collilineatus and C.d. terrificus from Southern Brazil. The possible relevance of
the observed venom variability among the tropical rattlesnake subspecies was assessed
by antivenomics using anti-crotalic antivenoms produced at Instituto Butantan and
Instituto Vital Brazil. The results revealed that both antivenoms exhibit impaired
immunoreactivity towards crotamine and display restricted (∼60%) recognition of PLA2
molecules (crotoxin and D49-myotoxins) from C. d. cascavella and C. d. terrificus venoms. This
poor reactivity of the antivenoms may be due to a combination of factors: on the one hand,
an inappropriate choice of the mixture of venoms for immunization and, on the other hand,
the documented low immunogenicity of PLA2 molecules. C. durissus causes most of the
lethal snakebite accidents in Brazil. The implication of the geographic variation of venom
composition for the treatment of bites by different C. durissus subspecies populations is
discussed.
© 2010 Elsevier B.V. All rights reserved.

1. Introduction heterodimeric PLA2 molecule exhibiting presynaptic β-neuro-


toxicity [20–22]. Crotoxin typically account for 70–90% of C.
Rattlesnakes (genera Crotalus and Sistrurus) represent a durissus venom toxins [23,24]. Crotalus durissus ssp. venoms
monophyletic clade of pitvipers which likely originated in also contain variable amounts of crotamine, a myonecrotic
the pine–oak, woodlands of the Sierra Madre Occidental in the toxin acting on voltage-sensitive Na+ channels [25,26]. Com-
north-central part of the Mexican Plateau several million years parative venomics of the Central American rattlesnake
after a single lineage of an Asian pitviper crossed to North C. simus and the South American C. durissus complex points
America via the Behring land bridge during the late Oligocene to neurotoxicity and lethal venom activities to rodents,
or early Miocene, 22–24 Mya [1–6]. Rattlesnakes dispersed associated to an increased concentration of neurotoxins
northward into North America and southward into South crotoxin and crotamine, as an adaptive paedomorphic trend
America [7,8] giving rise to the approximately 70 species and along Crotalus dispersal in South America [24]. Identifying the
subspecies (http://www.reptile-database.org), which are cur- molecular basis of adaptations in natural populations is an
rently discontinuously found from southern Canada to important yet largely unrealized goal in evolutionary biology
northern Argentina [9–11]. In Brazil, genus Crotalus is repre- [27–31]. Given the central role that diet has played in the
sented by a single species, the Neotropical rattlesnake Crotalus adaptive radiation of snakes [32], venom represents a key
durissus [12]. Biogeographical data suggest a relatively recent factor in the diversification of these animals. On the other
South American dispersal of C. durissus across a central trans- hand, adequate treatment of envenoming is critically depen-
Amazonian corridor during the middle Pleistocene (1.1– dent on the ability of parenteral-administered antivenoms to
1.0 Mya) followed by vicariance and dispersal into northeast- neutralize the lethal toxins reversing thereby the pathological
ern Brazil [12,13]. The assemblage of forms currently known as symptoms [33,34]. Hence, besides ecological implications,
the subspecies of the Neotropical rattlesnake constitute a set knowledge of the natural history and toxin composition of
of closely related parapatric forms, whose mutual relations venoms is of fundamental importance for the treatment of bite
and mechanisms of speciation remain largely elusive. Seven victims and in the selection of specimens for the preparation
subspecies of C. durissus are recognized in Brazil (C. d. dryinas, of venom pools for antivenom production [35,36]. This is
C. d. marajoensis, C. d. ruruima, C. d. trigonicus, C. d. terrificus, C. d. particularly relevant for highly adaptable and widely distrib-
cascavella, and C. d. collilineatus) [14] (Fig. 1). However, some uted species, in which intraspecific (ontogenetic, individual,
authors consider C. d. cascavella and C. d. collilineatus in the and geographic) venom variability represents a well documen-
synonymy of C. d. terrificus [12,15]. ted phenomenon [37–39] and a source of diversity of the
Species of the C. durissus complex pose a serious medical pathological effects of snakebites in different geographical
problem in many parts of their range, being responsible for regions [40,41]. Here we sought to characterize the venom
most of the lethal snakebite accidents in Brazil [14,16], where C. phenotypes of adult specimens of subspecies cascavella and
d. cascavella, C. d. collilineatus and C. d. terrificus display wide collilineatus of Brazilian C. durissus and neonate C. d. terrificus.
ecological and geographical distribution (Fig. 1). Venoms of The immunoreactivity of the anti-crotalic antivenoms pro-
these rattlesnakes are notorious for their ability to decouple duced at Instituto Vital Brazil (Niterói, Rio de Janeiro, Brazil)
neuromuscular transmission and for provoking myalgic and Instituto Butantan (São Paulo, Brazil) against bites of these
symptoms causing progressive paralysis [17,18]. Systemic taxa was investigated using an antivenomics approach. Our
neurotoxicity is frequently accompanied by rhabdomyolysis results reveal population-specific venom features and the lack
causing acute tubular necrosis and renal failure, which of immunoreactivity of the two antivenoms towards crota-
represents the primary cause of death [17,18]. Systemic mine, highlighting the need to revisit current immunization
neuro- and myotoxicity upon C. durissus ssp. bites are mainly protocols for producing an improved anti-crotalic antivenom
attributable to the high concentration of crotoxin [19], a displaying broad-ranging clinical use in Brazil.
1760 J O U R NA L OF PR O TE O MI CS 73 ( 20 1 0 ) 1 7 5 8– 1 7 7 6

Fig. 1 – Distribution and evolutionary trends among South American rattlesnake. (A) Geographic distribution of C. d. cascavella,
C. d. collilineatus and C. d. terrificus in Northern, Midwestern, and Southern Brazil, respectively. Symbols indicate the
approximate regions of origin of the snakes sampled in this work: ▲, C. d. cascavella; ■, C. d. collilineatus; ● and ♦, C. d. terrificus.
Pictures of C. d. collilineatus, C. d. cascavella and C. d. terrificus were generously provided by Renata S. Rodrigues, Breno
Handan (Nucleo Regional de Ofiologia e Animais Peçonhentos da Bahia (NOAP/UFBA)), and Rodolfo Capdevielle (http://www.
serpientes-snakes.com.ar), respectively. Intraperitoneal median lethal dose ranges (LD50) have been reported in the literature
[15,102] and are expressed in μg/kg. LNC (lethal neurotoxicity coefficient) is defined as the ratio between the average LD50 of the
venom and the neurotoxin (crotoxin + crotamine) concentration [24]. (B) The evolutionary trend toward increasing neurotoxicity
to mice among C. durissus ssp. venoms involves decrease of myotoxic D49-PLA2 concentration concomitantly to an enhanced
expression of crotoxin and crotamine along the dispersal axis of this taxa.

from neonates was obtained by aspiration from the fangs using


2. Experimental section an Eppendorf pipette. Crude venoms were centrifuged at low
speed to remove cells and debris, lyophilized, weighed on a
2.1. Venoms and antivenoms microbalance, and stored at − 20 °C until used. The anti-crotalic
antivenoms used for the antivenomic study were kindly
Crude venoms of adult Crotalus durissus terrificus were pooled provided by Instituto Butantan (But, São Paulo, Brazil, http://
from specimens collected in Juiz de Fora (Minas Gerais) www.butantan.gov.br) and Instituto Vital Brazil (IVB, Niterói,
(Southeastern population) and in the state of Rio Grande do RJ, Brazil; http://www.ivb.rj.gov.br). These antivenoms were
Sul (Southern population), and from the captive-born 12-weeks produced by hyperimmunization of horses with a pool of
old offspring of specimens from the Southern population, all venoms. Instituto Butantan uses a mixture of equal amounts
kept in the serpentarium of the Instituto Vital Brazil, Niterói-RJ, of C. d. terrificus and C. d. collilineatus venoms collected in
Brazil. Venom from C. d. collilineatus was pooled from adult Southeastern and Midwestern Brazil, in the states of São Paulo,
specimens collected in the state of Goiás, Brazil, and kept in Mato Grosso and Minas Gerais (Marisa Maria Teixeira da Rocha,
captivity in the serpentarium Bioagents of Batatais-SP, Brazil. Instituto Butantan, personal communication); the antivenom
Venom of C. d. cascavella was pooled from adult specimens manufactured at IVB was produced in horses using venom
collected in Bahia, Brazil and kept in the serpentarium of pooled from C. d. terrificus specimens from South-Eastern Brazil
Nucleo Regional de Ofiologia e Animais Peçonhentos da Bahia (Dr. Aníbal Melgarejo, Instituto Vital Brazil, personal commu-
(NOAP/UFBA), BA, Brazil. The venom from adult animals was nication). Both antivenoms comprise purified F(ab′)2 frag-
collected by snake biting on a parafilm-wrapped jar. Venom ments generated by pepsic digestion of ammonium sulphate-
J O U R NA L OF PR O TE O MI CS 7 3 ( 2 01 0 ) 1 7 5 8– 1 7 7 6 1761

precipitated IgGs. F(ab′)2 concentration was determined spec- evaluated by staining the nitrocellulose membranes with
trophotometrically using an extinction coefficient (ε) of 1.4 for Ponceau-S Red. Membranes were then destained in water,
a 1 mg/ml concentration at 280 nm using a 1 cm light path- incubated in 1% bovine serum albumin in 0.12 M NaCl, 0.04 M
length cuvette [42]. sodium phosphate, pH 7.2 (blocking buffer), for 30 min at room
temperature to block non-specific reactive sites, and incubat-
2.2. Isolation and characterization of venom proteins ed overnight at 4 °C with antivenom diluted 1:2000 (v/v) in
blocking buffer. After washing 4 times with blocking buffer
Venom proteins were separated by reverse-phase HPLC using containing 0.05% Tween-20, the membranes were incubated
a Teknokroma Europa C18 (0.4 cm × 25 cm, 5 mm particle size, for 2 h at room temperature with alkaline phosphatase-
300 Å pore size) column as described [24,35,36,43]. Protein conjugated anti-horse IgG (Sigma) diluted 1:2000, washed 4
detection was at 215 nm and peaks were collected manually times as above, and developed using ECL PLUS Western
and dried in a Speed-Vac (Savant). The relative abundances (% Blotting Detection Reagents (GE-Healthcare).
of the total venom proteins) of the different protein families in
the venoms were estimated from the relation of the sum of the 2.4. Antivenomics: immunodepletion of venom proteins by
areas of the reverse-phase chromatographic peaks containing polyvalent antivenoms
proteins from the same family to the total area of venom
protein peaks. In a strict sense, and according to the Lambert– We have coined the term “antivenomics” for the proteomic
Beer law, the calculated relative amounts correspond to the “% identification and quantitation of venom proteins escaping
of total peptide bonds in the sample”, which is a good estimate antivenom recognition [35,36,39,46–50]. Briefly, 2 mg of crude
of the % by weight (g/100 g) of a particular venom component. venom were dissolved in 70 μL of 20 mM phosphate buffer, pH
The relative contributions of different proteins eluting in the 7.0, mixed with 4 mg of But or IVB antivenom, and incubated
same chromatographic fraction were estimated by densitom- with gentle stirring overnight at 37 °C. 6 mg (350 μL) of rabbit
etry after SDS-PAGE separation. anti-horse IgG antiserum (Sigma) were then added and the
HPLC fractions were analyzed by SDS-PAGE (on 12 or 15% mixture was allowed to react for 2 h at 37 °C. Immunocom-
polyacrylamide gels) and N-terminal sequencing (using a plexes were precipitated by centrifugation at 13.000 ×g for
Procise instrument, Applied Biosystems, Foster City, CA, 30 min in an Eppendorf centrifuge, and the supernatant was
USA). Amino acid sequence similarity searches were per- analyzed by reverse-phase HPLC as above. Control venom
formed against the available databanks using the BLAST samples were subjected to the same procedure except that
program [44] implemented in the WU-BLAST2 search engine non-immune IgG instead of antivenom was included in the
at http://www.bork.embl-heidelberg.de. Molecular mass de- reaction mixture.
termination was performed by MALDI-TOF mass spectrometry
(using an Applied Biosystems Voyager-DE Pro™ instrument) 2.5. Generation of rabbit antibodies against C. d. terrificus
and electrospray ionization (ESI) mass spectrometry (using an venom
Applied Biosystems QTrap™ 2000 mass spectrometer). Protein
bands of interest were excised from Coomassie Brilliant Blue- To assess immunogenicity of crotamine, an experimental
stained SDS-PAGE gels and subjected to automated reduction, antiserum was raised in rabbits by subcutaneous injections of
alkylation, and in-gel digestion with sequencing grade porcine sublethal amounts of a mixture of crotalid venoms (to be
pancreatic trypsin (Promega). Doubly- or triply-charged ions of reported elsewhere), including crotamine-positive C. d. terrifi-
peptides selected from the MALDI-TOF mass fingerprint cus venom pooled from Southern Brazil specimens. First
spectra of the tryptic digests were sequenced by CID-MS/MS injection comprised 100 μg venom in 100 μL of PBS (20 mM
using an Applied Biosystem's QTrap 2000. CID spectra were phosphate, 135 mM NaCl, pH 7.3) emulsified with an equal
interpreted manually or using a licensed version of the volume of Freund's complete adjuvant. Booster injections
MASCOT program (http://www.matrixscience.com) against a comprising increasing amounts (200–500 μg) of immunogen
private database containing viperid protein sequences depos- emulsified in Freund's incomplete adjuvant were adminis-
ited in the SwissProt/TrEMBL database plus the previously tered every 2 weeks for a period of 2–3 months (depending on
assigned peptide ion sequences from snake venomics projects the titer of the antiserum determined by a standard ELISA
carried out in our laboratory. MS/MS mass tolerance was set to procedure). Terminal cardiac blood collection, done by intra-
±0.6 Da. Carbamidomethyl cysteine and oxidation of methio- cardiac puncture performed under general anesthesia, was
nine were fixed and variable modifications, respectively. approved by the IBV's Ethics Commission. The IgG fraction
was purified by ammonium sulphate precipitation followed by
2.3. Western blot analysis affinity chromatography on Sepharose-Protein A (Agarose
Bead Technologies, Tampa, FL, USA) following the manufac-
For assessing the immunoreactivity of the IVB and But anti- turer's instructions. For Western blot analysis, purified IgG
crotalic antivenoms towards C. durissus venom proteins by was used at a concentration of 10 μg/ml. For antivenomics,
Western blot analysis, the reverse-phase HPLC chromato- 1 mg of C. d. terrificus venom in 300 μlL of 0.2 M phosphate, pH
graphic fractions were electrophoresed in SDS-PAGE (12 or 7.0, was incubated with 10 mg of rabbit IgG antibodies
15%) polyacrylamide gels under non-reduced conditions overnight at room temperature and with gentle stirring. IgG–
followed by electrotransfer to Hybond™-P membrane (GE- antigen complexes were immunoprecipitated with Agarose-
Healthcare) [45] using a Bio-Rad Semi-Dry minitransfer cell Protein-A (ABT), and the supernatant was analyzed by
operated at 200 V during 120 min. Transfer efficiency was reverse-phase HPLC as above.
1762 J O U R NA L OF PR O TE O MI CS 73 ( 20 1 0 ) 1 7 5 8– 1 7 7 6

belonging to 8 protein families (Fig. 4, Tables 1 and 2). PLA2


3. Results and discussion molecules, particularly neurotoxic crotoxin isoforms, which
accounts for 70–80% of the total venom proteins, constitute
3.1. The venom proteomes of C. d. collilineatus and C. d. the major toxin of both taxa. Disintegrins, thrombin-like and
cascavella gyroxin-like serine proteinases, CRISP, svVEGF, L-amino acid
oxidase, C-type lectin-like convulxin, and PIII-SVMPs all
The proteome of venom pooled from adult C. d. collilineatus represent minor components, none of which is present in
(Fig. 2A) and C. d. cascavella (Fig. 3) specimens share toxins concentration higher than 2% of the total venom proteins

Fig. 2 – Characterization of the venom proteome of Crotalus durissus collilineatus. Panels A and B display, respectively,
reverse-phase HPLC separations of the proteins of pooled and individual venoms. Two milligrams of total venom proteins were
applied to a Europa Protein-300 C18 column, which was then developed with the following chromatographic conditions: isocratically
(5% B) for 10 min, followed by (5–15% B) for 20 min, (15–45% B) for 120 min, and (45–70% B) for 20 min. Fractions were collected
manually and submitted to N-terminal sequencing and molecular determination by SDS-PAGE, ESI and MALDI mass spectrometry.
The results are shown in Table 1. Inset panels: Coomassie blue-stained SDS-AGE showing the protein composition of the
reverse-phase HPLC-separated venom protein fractions displayed in the respective panel and run under non-reduced (upper panels)
and reduced (lower panels) conditions. Molecular mass markers (in kDa) are indicated at the side of each gel. Selected protein bands
were excised and characterized by mass fingerprinting and CID-MS/MS of selected doubly- or triply-charged peptide ions (Table 1).
J O U R NA L OF PR O TE O MI CS 7 3 ( 2 01 0 ) 1 7 5 8– 1 7 7 6 1763

Fig. 3 – Characterization of the venom proteome of Crotalus durissus cascavella. The proteins contained in two milligrams of
whole dried pooled venom were separated by reverse-phase HPLC and analyzed as described in the legend of Fig. 2. The results
are shown in Table 1. Inset, Coomassie blue-stained SDS-PAGE of the reverse-phase HPLC protein fractions run under
non-reduced (upper panels) and reduced (lower panels) conditions. Molecular mass markers (in kDa) are indicated at the side of
each gel. Selected protein bands were excised and characterized by mass fingerprinting and CID-MS/MS of selected doubly- or
triply-charged peptide ions (Table 1).

(Fig. 4, Table 2). Shared venom components between C. d. pools of C. d. collilineatus and C. d. cascavella is the large amount
collilineatus and C. d. cascavella are apparently identical as of the myotoxin crotamine in the former, and the increased
judged by reverse-phase HPLC elution profile, N-terminal concentration of acidic D49-PLA2 [C9E7C4] (fraction 14) in the
sequencing, SDS-PAGE, MALDI-TOF mass fingerprinting and latter (Fig. 4, Tables 1 and 2). Notwithstanding these differ-
MS/MS analysis, and are identified in the respective HPLC ences, C. d. collilineatus and C. d. cascavella share about 90% of
separations and in Table 1 with the same numbering. their venom proteomes, and closely resemble that of C. d.
However, the relative abundance of these proteins varied terrificus [24, but see also Fig. 5], supporting the view that, from
among the venoms, each venom comprising some unique a venomics perspective, these three taxa might be considered
components. The most notable departure between the venom geographic variations of the same species [12,15]. However, a

Fig. 4 – Overall protein composition of the venoms of C. d. collilineatus and C. d. cascavella. Pie charts of relative abundance of
different toxin families in the pooled (A) and individual (B) venom of C. d. collilineatus and the pooled venom of C. d. cascavella (C).
svVEGF, snake venom vascular endothelial growth factor; CRISP, Cysteine-rich secretory protein; LAO, L-amino acid oxidase;
CTL, C-type lectin-like protein; PIII-SVMP, snake venom metalloproteinase from class III; SerProt, serine proteinase; PLA2,
phospholipase A2. Details of the individual proteins are shown in Table 1 and the percentages of the different toxin families in
the venoms are listed in Table 2.
1764 J O U R NA L OF PR O TE O MI CS 73 ( 20 1 0 ) 1 7 5 8– 1 7 7 6

Table 1 – Assignment of the reverse-phase fractions from the venom pools of Crotalus durissus collilineatus and Crotalus
durissus cascavella, isolated as in Figs. 2A and 3A, to protein families by N-terminal Edman sequencing, mass spectrometry,
and collision-induced fragmentation by nESI-MS/MS of selected peptide ions from in-gel digested protein bands separated
by SDS-PAGE (insets in Figs. 2A and 3A). X, Ile or Leu; Z, pyrrolidone carboxylic acid. Cp, propionamide cysteine. Unless
other stated, for MS/MS analyses, cysteine residues were carbamidomethylated; Molecular masses of native proteins were
determined by electrospray ionization (±0.02%) or MALDI-TOF (±0.2%) mass spectrometry. Apparent molecular masses were
determined by SDS-PAGE of non-reduced (■) and reduced (▼) samples.
HPLC N-terminal Molecular Peptide MS/MS-derived Protein family
fraction mass ion sequence

m/z z

1–3 YKQCHKKGGHCFPKE 4884 Da 610.6 2 ICLPPSSDFGK Myotoxin Crotamine 1 [P01475]


4982 Da Myotoxin Crotamine isoform
4 (R)IECDCGSIENPCCYA 7305 Da 812.6 2 NDDTCpTGQSADCpPR Disintegrin [COL2T8] 408–474
5 (R)IECDCGSIENPCCYA 7519 Da 812.6 2 NDDTCpTGQSADCpPR Disintegrin [COL2T8] 408–476
7721 Da ∼ Disintegrin [COL2T8] 408–478
(R)IECDCGSIENPCCYA 7593 Da ∼ Disintegrin [COL2T8] 408–477
IECDCGSIENPCCYAT 7364 Da Disintegrin [COL2T8] 408–476
6–9 SSYGCYCGAGGQGWP 9■/▼ kDa Crotoxin acid chain [P08878]
A-chain (38–83)-SS-B-chain
(84–124)-SS-C-chain (125–138)
7 Blocked 16.5■/ 831.5 2 LTGCDPTTDVYTYR Crotoxin acid chain [P08878]
7▼ kDa
780.3 2 CCFEHDCCYAK
10 SLLQFNKMIKFETRK 14,197 Da 871.8 2 GTWCEEQICECDR ∼ Crotoxin basic chain
[P62022, I109]
633.7 2 YGYMIYPDSR
11 a HLLQFNKMIKFETRK 14,248 Da Crotoxin basic chain [P0CAS2]
b NLLQFNKMIKEETGK 14,143 Da ∼ Presynaptic neurotoxic PLA2
[AAB34413]
12 a HLLQFNKMIKFETRK 14,187 Da 649.6 2 YGYMFYPDSR Crotoxin basic chian 1 [P62022]
450.3 2 HLLQFNK
871.5 2 GTWCEEQICECDR
592.8 2 WDIYPYSLK
720.3 3 NAIPFYAFYGCYCGWGGR
b SLLQFNKMIKFETRK 14,284 Da ∼ Crotoxin basic chain [P62022]
c NLLQFNKMIKEETGK 14,143 Da ∼ Presynaptic neurotoxic PLA2
[AAB34413]
13 a Blocked 25,733, 472.2 2 PVCVTVXR svVEGF [∼ ACN22040]
25,835 Da
682.4 2 XEVMQFTEH(249.2)
750.3 3 (374.3)VSILDEYPSEVAHLFR
706.3 2 (342.2)PFMEVYER
b SVDFDSESPRKPEIQ 23 kDa▼ 569.7 2 SVDFDSESPR CRISP
c SLVEFETLMMKIAGR 14 kDa▼ 831.4 2 LTGCDPTTDVYTYR ∼ Crotoxin acid chain [P08878]
14, 15 SLLDFEMMIIKVAKK 13,836 Da 752.8 2 CCFVHDCCYGK ∼ acidic D49–PLA2 [C9R7C4]
511.2 2 QFPAENCR
670.6 2 SLLDFEMMIIK
836.8 2 QTCECDGVAAVCFR
13–18 SLLDFEMMIIKVAKK 14,245 Da 518.2 2 QFPAENCpR ∼ acidic D49–PLA2 [C9E7C4]
15 VIGGDECNINEHNFL 28 kDa▼ 728.4 2 THFLIYVGVHDR ∼ Serine porteinase Gyroxin-like
B1_4
661.4 2 HILIYVGVHDR [B0FXM2]
568.8 2 SVQFDKEQRR
16 VIGGDECNINEHNFL 28 kDa▼ 728.4 2 THLIYVGVHDR ∼ Serine proteinase Gyroxin-like
B1_4
874.3 2 ILCAGVLEGGIDTCNR [B0FXM2]
773.8 3 NNEHIAPLSLPSSPPSVGSVCR
568.8 2 SVQFDKEQRR
647.8 2 XNXXDYEVCR
17 GLHCPSDWYYYDQH 110■/ 534.3 2 WFVASCIGK C-type lectin convulxin-α
14–16▼ kDa [Ο93426]
GFCCPSHWSSYDRY 774.3 3 GLHCPSDWYYYDQHCYR
829.8 2 GFCCPSHWSSYDR C-type lectin convulxin-β
[Ο93427]
849.4 2 STFFWIGANNIWNK
809.9 3 TFDNQWLSAPCSDTYSFVCR
J O U R NA L OF PR O TE O MI CS 7 3 ( 2 01 0 ) 1 7 5 8– 1 7 7 6 1765

Table 1 (continued)
HPLC N-terminal Molecular Peptide MS/MS-derived Protein family
fraction mass ion sequence

m/z z
■/▼
18 a Blocked 115 kDa 577.9 2 EGNHYGYCRKEQNTK PIII-metalloproteinase [C5H5D1]
b N.D. 58 kDa▼, ■ 485.6 2 VQVHFNAR L-amino acid oxidase [∼ P56742]
519.3 2 FWEDDGIR
532.3 2 NPLEECFR
583.3 2 IKFEPPLPPK
682.6 2 SAAQLYVESLRK
757.8 2 ETDYEEFLEIAR
619.2 2 SAAQLYVESLR
935.2 2 NNPGLLEYPVKPSEEGK
612.2 2 DWYANLGPMR
557.8 2 VIEIQQNDR
c N.D. 28 kDa▼, ■
728.4 2 THFLIYVGVHDR Serine porteinase Gyroxin-like B1_4
661.4 2 HILIYVGVHDR [B0FMX2]
15–18 N.D. 15 kDa▼, ■
649.6 2 YGYMFYPDSR Crotoxin basic chain 1[P62022]
871.5 2 GTWCEEQICECDR
753.3 2 CCFVHDCCYGK
19 VVGHPCNINEHRSL 28 kDa▼ Serine proteinase
20 VIGGHPCNINEHNFL 28 kDa▼ Serine proteinase

thorough proteomic investigation of the 2DE-separated the total toxins of C. d. collilineatus pooled venom (Table 2).
venom proteome of C. d. terrificus [51] identified eight 5′- However, this toxin was not found in the individual venom
nucleotidase isoforms, 2 phosphodiesterase spots, and one (Table 2), indicating the existence of crotamine-positive and
glutaminyl cyclase, which comprised respectively 7–8%, 1.9%, crotamine-negative C. d. collilineatus specimens. The transla-
and 1% of total venom proteins, but have been detected tional rate of the crotamine mRNA has not been studied, and
neither in previous reverse-phase separation of C. d. terrificus thus it remains uncertain whether the specimen used for the
[24] nor in C. d. collilineatus and C. d. cascavella venoms transcriptomic analysis belonged to a crotamine-positive or to
[this work]. Furthermore, Georgieva et al. [51] did not found a crotamine-negative population. In the few instances in
L-amino acid oxidase and the C-type lectin-like convulxin in which transcriptomics and proteomics databases have been
C. d. terrificus venom, whereas these proteins are clearly compared [52–54], a low degree of accordance has been
present in the C. d. collilineatus and C. d. cascavella venoms reported, indicating that the cDNA libraries lacked many
sampled in this work (Table 2), and have been reported to transcripts encoding venom-expressed proteins. On the other
comprise about 3.6% and 0.2% of the venom proteome of C. d. hand, the occurrence of non-translated messengers point
terrificus in a previous work [24]. Our results also show the out to transcripts exhibiting an individual or a temporal
presence of PIII-metalloproteinase(s) in the three C. durissus expression pattern over the life time of the snake [37].
subspecies [24, Table 2] although this toxin family was not The cocktail of toxins present in C. durissus ssp. venoms
detected in the 2DE-separated venom proteome of C. d. explains the clinical picture observed upon envenomations in
terrificus [51]. In addition, C. d. collilineatus and C. d. cascavella, Brazil. The serious neurotoxic symptoms are attributable to the
but not C. d. terrificus [24,51], contain CRISP (1.1–2.6% of total high amounts of the β-neurotoxin crotoxin [19–22,55]. Crotoxin
reverse-phase-separated venom proteins) (Table 2). As a has also been recognized as the main component responsible
whole, these evidences indicate the occurrence of individual for acute nephrotoxicity [56,57], although the myonecrotic
or geographic variability among C. durissus spp. venoms. activity of crotamine [25,26,58] may also contribute to rhabdo-
In line with the proteomic outcome reported here, a recent myolysis and acute kidney failure. Other potentially life-
analysis of toxin-encoding ESTs from a C. d. collilineatus cDNA threatening clinical effects encountered throughout the
library [23] identified crotoxin as the most expressed tran- range of C. durissus such as hemostatic disturbances, myocar-
script, comprising 88% of all toxin-coding sequences. Minor dial damage, hypotension and shock [18,19] may involve the
components (accounting for less than 3% of the total venom combined or synergistic effect of the less abundant toxins.
proteins) found in both, transcriptome and proteome include Hence, RGD disintegrin [COL2T8] [23] (HPLC fractions 4, 5)
svVEGF, serine proteinases (including gyroxin), the C-type blocks platelet aggregation; thrombin-like serine proteinases
lectin-like convulxin, and SVMPs (Table 2). However, low (i.e. gyroxin [B0FXM2] found in fractions 15, 16, and 18–20) may
abundant transcripts encoding nerve growth factor, cardio- produce hypofibrinogenemia and incoagulable blood [59,60];
toxin, angiotensin-converting enzyme inhibitor, C-natriuretic gyroxin, in addition, induces the neurological syndrome of
peptide, ohanin, and PLA2 inhibitor, which comprised 2.5% of “barrel rotation”, characterized by opisthotonos and rotations
toxin-coding ESTs, are not mirrored in the venom proteome around the long axis of the animal [61]. The acidic D49-PLA2
(Tables 1 and 2). In addition, crotamine, which was identified [C9E7C4] [62] eluted in HPLC fractions 13–18 may induce local
in the transcriptome as a singleton [23], represents 21% of tissue damage associated to inflammation [63]. Fraction 18a
1766 J O U R NA L OF PR O TE O MI CS 73 ( 20 1 0 ) 1 7 5 8– 1 7 7 6

corresponds to crotastatin-1 [C5H5D1], a vascular apoptosis- venom proteins in the Southern population but only 2% in the
inducing (VAP) dimeric PIII-SVMP [64]. The L-amino acid venom from the Southeastern pool. Venom pooled from
oxidase eluted in fraction 18b was previously isolated by neonates born from mothers from the Southeastern popula-
Toyama and co-workers [65] from C. d. cascavella venom tion were all crotamine-negative (Fig. 5C). Except for the
and exhibited strong platelet aggregation activity. Tetrameric absence of crotamine, neonate and adult snakes from the
C-type lectin-like convulxin [O93427] (HPLC fraction 17) has same geographical pool both express type II venoms exhibit-
been also characterized as a potent platelet aggregation agent ing highly similar toxin profiles, thus essentially supporting
[66–68]. the predicted trend. Nonetheless, serine proteinases 15 and 16
showed higher expression level in venom pooled from
3.2. Change in the PLA2 profile of C. d. cascavella and neonates (∼ 12%) (Fig. 5C) than in venom from adults (∼ 0.5–
C. d. collilineatus venoms: an adaptive specialization 4%) (Fig. 5B and A, respectively). This may explain the higher
to neurotoxicity coagulant and fibrinolytic activities reported in the venom of
newborn versus adult C. d. terrificus [71].
Venoms of Neotropical Crotalus subspecies belong to one of Fig. 1 shows the geographic distribution of C. d. cascavella,
two distinct phenotypes, which broadly correspond to type I C. d. collilineatus and C. d. terrificus along with the estimated
(high levels of SVMPs and low toxicity, LD50 > 1 μg/g mouse LNC for their venoms, calculated using LD50 values reported in
body weight) and type II (low metalloproteinase activity and the literature [15]. The data showing a clear tendency of C.
high toxicity, LD50 < 1 μg/g mouse body weight) venoms durissus ssp. venoms towards decreasing LNC values from
defined by Mackessy [69,70]. In a previous comparative Northeastern through Southern Brazil, is in agreement with
venomic study, we reported the occurrence of ontogenetic the hypothesis that neurotoxicity to mammals represents the
change from type II (neurotoxic) to type I (hemorrhagic) key axis along which overall venom toxicity has evolved
venom in Central American C. simus, and hypothesized that during the southward dispersion of C. durissus ssp. in South
neurotoxicity represented a paedomorphic trend and an America [24]. In line with this view, Powell and Lieb [72] have
adaptive trait along the dispersal of Crotalus durissus ssp. in predicted that the extremely high neurotoxicity exhibited by
South America [24]. This hypothesis predicted i) the absence of North American rattlesnakes represents a transitory popula-
ontogenetic change in venoms of Crotalus durissus ssp., and tional phenomenon associated with novel prey bases. The
ii) a gradual decreasing of the “lethal neurotoxicity coefficient” venoms of these species contain Mojave neurotoxin [acid
(LNC, defined as the ratio between the average LD50 of the chain, P18998; basic chain, P62023], the North American
venom and its relative amount of neurotoxins) along the homolog of South American crotoxin [73]. Niche colonization
dispersal route of Crotalus southward of the Amazon basin. by development of venom neurotoxicity has also been
The first prediction, lack of ontogenetic venom variation, was described in European Viperid snakes. The major toxic
tested through comparison of the toxin profiles of venoms component of the V. a. meridionalis venom is vipoxin
pooled from neonate and adult specimens of C. d. terrificus [P14420], a heterodimeric postsynaptic PLA2 neurotoxin
(Fig. 5). It was found that venoms pooled from adult snakes [74,75]. Since 1992, the Marseille Poison Center has regularly
from the Southeastern (Fig. 5A) and Southern (Fig. 5B) observed the occurrence of cases of human envenomation
populations strongly depart in their crotamine content. The after Vipera ammodytes ammodytes bites in south-east France
relative abundance of this toxin averaged 22% of the total involving unexpected neurological signs [76]. In France,
neurological symptoms had previously only been described
in cases of envenomation by Vipera aspis zinnikeri located
mainly in the south-west of France. Venom PLA2 profiling,
Table 2 – Overview of the relative occurrence of proteins genetic analyses, and epidemiological survey of snake bites in
(in percentage of the total HPLC-separated proteins) of the
various regions of France all indicate that the neurotoxic V. a.
different families in the venoms of C. durissus subspecies
collilineatus and cascavella. ammodytes population may have originated from a “crypto-
neurotoxic” V. a. zinnikeri population that may have colonized
Proteins family % of total venom proteins
northern areas from the ice refuges of the southwest of France,
C. d. collilineatus C. d. cascavella by moving along the rivers, which constitute thermal corridors
[77].
Pooled Individual Pooled
C. d. cascavella venom contains, in addition to neurotoxic
Crotamine 20.8 – – PLA2, a considerable amount (18% of the total venom proteins)
Disintegrin 0.5 0.4 0.2 of the acid D49-PLA2 [C9E7C4] (HPLC peak 14 in Fig. 3; Table 2).
PLA2 72 90.9 90.6
The amount of this myotoxin is much reduced in the pooled
f Crotoxin
Other PLA2s
67.4
4.6
79.6
11.3
72.5
18.1
venoms of C. d. collilineatus (4.6%) (Fig. 2; Table 2), and C. d.
Serine Proteinase 1.9 0.5 1.2 terrificus (2.1%). This observation, which is in line with
CRISP 1.8 1.6 0.9 previous biochemical evidences showing that C. durissus
svVEGF 2.1 2.6 1.1 cascavella venom presented the highest PLA2 activity, whereas
L-amino acid 0.5 2.6 < 0.1 C. durissus collilineatus the lowest [15]. We interpret the
oxidase decrease of non-neurotoxic PLA2 as part of the venom
C-type lectin-like <0.1 < 0.1 < 0.1
specialization trend of C. durissus ssp. toward neurotoxicity
PIII-Zn2+- 0.4 2.3 < 0.1
metalloproteinase
during the colonization of new ecological niches on their
southward dispersion. A prediction of this view is that the
J O U R NA L OF PR O TE O MI CS 7 3 ( 2 01 0 ) 1 7 5 8– 1 7 7 6 1767

venom from Paraguayan and North Argentinian populations crotic toxin with β-defensin scaffold (PDB accession code
of C. d. terrificus may follow the trend toward lower levels of 1Z99) targeting voltage-sensitive Na+ channels, inducing
myotoxic D49-PLA2 in favor of an increase in the concentra- thereby skeletal muscle membrane depolarization [78], may
tion of crotoxin. also represent an adaptive trait. Crotamine-positive and
crotamine-negative populations were described more than
3.3. The trend toward increasing concentration and 50 years ago [79,80]. Now, we show an increasing expression
frequency of crotamine in C. d. durissus populations from of crotamine among C. durissus ssp. coincident with the
northeastern Brazil to northern Argentina suggests an direction of the dispersal of this taxa. Thus, the proteomic
adaptive role for this myotoxin analysis of pooled venom from C. d. cascavella (Fig. 3) corro-
borates the reported absence of crotamine in rattlesnakes
The outcome of the present investigation suggests that in from northeastern Brazil (Fig. 1) [15], and the occurrence of
South America C. durissus crotamine, a small basic myone- crotamine-positive and crotamine-negative populations of

Fig. 5 – Lack of ontogenetic venom variation in C. d. terrificus. The venom proteomes of adult C. d. terrificus from Southeast
(A) and Southern (B) Brazil, and from the pooled venom of captive-born 12-weeks old offspring of specimens from the Southern
population (C) were separated by reverse-phase HPLC and analyzed as described in the legend of Fig. 2. Peak numbering
corresponds to proteins listed in Table 1.
1768 J O U R NA L OF PR O TE O MI CS 73 ( 20 1 0 ) 1 7 5 8– 1 7 7 6

Figure 5 (continued).

C. d. collilineatus (Fig. 2) and C. d. terrificus (Fig. 5) in midwestern Caatinga region) is quite different from that of C. d. collilineatus
and south Brazil, respectively (Fig. 1). and C. d. terrificus (semideciduous forest, the Cerrado region).
Schenberg [79] reported an increase in the number of In addition, the feeding habits of C. d. cascavella consists on
crotamine-positive rattlesnakes to the west of the states of lizard, birds and small mammals, whereas C. d. collilineatus
São Paulo and Paraná, until a region was reached where only and C. d. terrificus feed exclusively on rodents from birth
crotamine-secreting snakes were found [80]. Furthermore, [87,88]. The adaptive value of crotamine may relay in a rapid
C. d. terrificus from Northern Argentina and Paraguay appear to paralyzation of rodents, thus securing the meal and avoiding
comprise genetically pure crotamine-secreting populations the risk of injury to the snake. Supporting this view,
[81]. This geographical distribution indicates the Mendelian crotamine has been shown to induce a potent analgesic effect
character of crotamine secretion [79]. Crotamine-like proteins when injected (s.c. and i.p.) or administered orally into mice
have evolved convergently in the venom of distant lineages, [89].
such as lepidosaurs and monotremes [82], indicating that
coopting these β-defensin proteins seems to be a common 3.4. Assessing the immunodepleting capability of
strategy for evolving venoms. In Crotalus, crotamine evolved Brazilian anti-crotalic antivenoms against the venoms of
in the venom gland by recruitment, duplication, and acceler- Crotalus durissus subspecies using antivenomics
ated evolution of its paralogous pancreatic crotasin gene [83].
The gene coding for crotamine (Crt-p1) has been located to the Given the wide distribution of C. durissus subspecies in Brazil,
end of the long arm of chromosome 2 of crotamine-positive and the occurrence of notable geographic venom variations,
specimens [84]. The crotamine gene exists from 1 to 32 we applied an antivenomics approach [35,36,39,46–50] to
copies per haploid genome, and the concentration of crota- investigate the in vitro immunoreactivity of the anti-crotalic
mine in the venom appears to be positively correlated with antivenoms produced at Instituto Vital Brazil and Instituto
the number of gene copies [85]. The variability in the copy Butantan. These two institutions, together with Centro de
number of crotamine genes might be explained by its Produção e Pesquisa de Imunobiológicos (Secretaria de Estado
subtelomeric location resulting in accelerated rate of dupli- da Saúde do Paraná), and Fundação Ezequiel Dias (FUNED,
cation and rearrangement [85,86]. The increased frequency of Secretaria de Estado de Saúde de Minas Gerais), form part of
crotamine-positive populations from northeastern Brazil to the network of institutions that manufacture antivenoms for
northern Argentina, suggests an adaptive value for crotamine. the Ministério da Saúde do Brasil. The Butantan antivenom
However, the mean LD50 values of crotamine for mice, 6.9 mg/ (But) efficiently immunodepleted serine proteinases, L-amino
kg (0.07–32.8 mg/kg, depending on the method used in acid oxidase, SVMPs and C-type lectin-like molecules from
determining lethality, such as mouse strain, site of inocula- the venom of adult (Fig. 6A) and juvenile (Fig. 6C) C. d.
tion, etc.) [86], is two orders of magnitude higher than the LD50 terrificus. However, this antivenom lacks immunoreactivity
of crotoxin (60–110 μg/kg). Thus, fixation of crotamine in the towards crotamine and disintegrin molecules (Fig. 6A) and
venom proteome concomitantly with C. durissus ssp. dispersal exhibits restricted (∼ 60%) immunodepletion ability of PLA2
in South America may reflect an adaptation to different molecules (crotoxin and D49-myotoxin) from C. d. terrificus
ecological niches and prey availability. Relevant to this (Fig. 6A) and C. d. cascavella (Fig. 7A) venoms. The But
argument, the habitat of C. d. cascavella (dry thorn scrub, the antivenom also showed negligible immunodepleting activity
J O U R NA L OF PR O TE O MI CS 7 3 ( 2 01 0 ) 1 7 5 8– 1 7 7 6 1769

towards serine proteinase 19 from C. d. cascavella (Fig. 7A). low immunogenicity of crotamine. The first possibility is
The immunodepleting ability of the IVB antivenom (Figs. 6B supported by the fact that Instituto Butantan uses a mixture
and 7B) did not significantly depart from that of the But of C. d. terrificus and C. d. collilineatus venoms collected in
antivenom. However, in Western blot analysis, the IVB crotamine-negative regions of Southeastern and Midwestern
antivenom, but not the antivenom from Instituto Butantan, Brazil. On the other hand, the low amount of crotamine in the
showed very weak immunorecognition of crotamine (Fig. 8). venom pooled from C. d. terrificus specimens from Southeast-
The distinct affinity of IVB and But antivenoms against ern Brazil (Fig. 5A) may explain the very weak immunor-
crotamine may be due to a combination of i) the different ecognition capability of the IVB antivenom toward crotamine.
mixtures of venoms used for hyperimmunizing horses and ii) On the other hand, the failure of snake antivenoms to

Fig. 6 – Immunodepletion of proteins from C. d. terrificus venoms by Brazilian anti-crotalic antivenoms. Panels A and B display,
respectively, reverse-phase separations of the proteins from pooled venom of adult C. d. terrificus recovered after antivenomic
treatment of the crude venom with the anti-crotalic antivenoms manufactured in Instituto Butantan (But) and Instituto Vital
Brazil (IVB). Panel C, reverse-phase HPLC of the non-immunodepleted proteins from the pooled venom of neonate C. d. terrificus
after incubation with antivenom from Instituto Butantan.
1770 J O U R NA L OF PR O TE O MI CS 73 ( 20 1 0 ) 1 7 5 8– 1 7 7 6

Figure 6 (continued).

recognize specific low molecular components of the venom Crotalus durissus ssp. [93], and the case fatality rate ranged
used for hyperimmunization, predominantly PLA2 proteins from 0.8% to 2.53% [94], thus constituting a relevant public
and α-neurotoxins has been also reported [46–49,90–92]. The health issue. The anti-crotalic antivenom produced at Insti-
possibility that lack of immunoreactivity against crotamine tuto Butantan (But) (São Paulo) against C. d. terrificus venom
was due to weak immunogenicity of the toxin was investi- shows a very high effectiveness in the neutralization of the
gated by immunizing rabbits against crotamine-positive C. d. lethal, myotoxin, and neurotoxic effects of venoms of C.
terrificus venom. The results, shown in Fig. 9, clearly durissus subspecies [63]. The anti-crotalic antivenom manu-
demonstrate that crotamine is able to induce a strong factured at Instituto Vital Brazil (IVB) (Niterói, Rio de Janeiro)
immune response and that the purified rabbit anti-crotamine has also been reported to neutralize the lethal activity of C. d.
IgG antibodies efficiently immunoprecipitate this toxin. terrificus venom [95]. This activity is mainly attributable to
crotoxin-induced neurotoxicity [16,17], and monospecific anti-
3.5. Concluding remarks and perspectives crotoxin antibodies neutralize the lethal activity of C. d.
terrificus [96] and C. d. cascavella [97] venoms. Our antivenomic
This work supports a previous report pointing to neurotoxicity results indicate that both, the But and the IVB antivenoms
as an adaptive trend along Crotalus dispersal in South exhibit suboptimal crotoxin immunodepleting activity. This
America [24]. In addition, results presented here indicate does not seem to be due to low immunogenicity of crotoxin, as
that this venom specialization trend occurred concomitantly evidenced by the fact that an experimental antiserum against
with decrease of non-neurotoxic D49-PLA2 concentration and the venom of C. d. terrificus developed in rabbits in our
with an enhanced expression of crotamine among C. durissus laboratory is capable of quantitatively immunoprecipitate
ssp. coincident with the direction of the dispersal of this taxa. the neurotoxin (Fig. 9).
The increased frequency of crotamine-positive populations Envenoming by C. durissus is also frequently associated
from northeastern Brazil to northern Argentina, suggests an with haemostatic disorders, likely due to the action of
adaptive value for crotamine. Besides ecological and taxono- thrombin-like enzymes [98]. Our results indicating an im-
mical implications, the identification of evolutionary trends, paired capability of the two Brazilian anti-crotalic antivenoms
as reported here, may have an impact in snakebite manage- sampled to immunodeplete serine proteinase 19 from the
ment. Hence, adequate treatment of snakebite envenoming is venom of C. d. cascavella (Fig. 7) should raise some concern
critically dependent on the parenteral administration of an among antivenom producers to try to improve this activity.
appropriate antidote that neutralizes the venom-induced However, the most unexpected and disturbing outcome of our
pathology, and the antivenom design requires a careful work is the absolute ineffectiveness of both, the But and IVB,
consideration of the venom mixtures to be used in the antivenoms to immunorecognize crotamine. Although from
immunization protocol [34,36]. its high LD50, crotamine seems not represent a life-threaten-
Accidental envenomations by subspecies of C. durissus are ing toxin, its role in human envenomation has not been
usually serious and frequently fatal in the absence of adequate entirely clarified. Crotamine produces muscle contracture of
treatment. In Brazil, 9% of the 26,688 snakebite cases reported peripheral origin, as well as spastic paralysis of skeletal
to have occurred between 2000 and 2007 were inflicted by muscle in mammals [99,100], and its reported myolytic
J O U R NA L OF PR O TE O MI CS 7 3 ( 2 01 0 ) 1 7 5 8– 1 7 7 6 1771

Fig. 7 – Immunodepletion of proteins from C. d. cascvavella venom by Brazilian anti-crotalic antivenoms. Panels A and B display,
respectively, reverse-phase separations of the proteins from pooled venom of adult C. d. cascavella recovered after antivenomic
treatment of the crude venom with the anti-crotalic antivenoms manufactured in Instituto Butantan (But) and Instituto Vital
Brazil (IVB).

activity may be consequence of impairing voltage-gated As a whole, our data suggest that the current immunization
sodium channels of skeletal muscle sarcolemma, and of protocols need to be revised in order to generate an improved
lysosomal membrane permeabilization leading to activation anti-crotalic antiserum. Optimization of the dose-efficacy of
of calcium-dependent proteases [101]. Crotamine is able to the antivenom will result in the need for lower volumes to
induce an immune response in rabbits, and therefore the effect treatment, with a consequent decrease in the risk of
negligible titer of the But and IVB antivenoms against serum-sickness and anaphylactic adverse effects. Venom
crotamine may be due to an unfortunate choice of the venoms phenotyping by reverse-phase HPLC may be considered to
(from crotamine-negative populations) used in the immuni- select the set of venoms containing all desired immunogens.
zation mixture. The increasing frequency of crotamine in Our antivenomic approach is simple and easy to implement,
southern populations of C. d. terrificus should be reason enough and may thus represent a useful proteomic tool for monitoring
to warn about the need to develop an antivenom capable of and quantifying the onset of the hyperimmunized animal's
neutralizing this toxin. immune response during antivenom production.
1772 J O U R NA L OF PR O TE O MI CS 73 ( 20 1 0 ) 1 7 5 8– 1 7 7 6

Fig. 8 – Western blot analysis. Crotamine isolated by reverse-phase HPLC from pooled C. durissus collilineatus venom (lane 1),
and the proteins from pooled C. d collilineatus venom (lane 2); pooled C. durissus terrificus venom (lane 3); individual C. d.
collilineatus venom (lane 4), and pooled C. d. cascavella venom were separated by SDS-PAGE (left panel), electroblotted onto
nitrocellulose membrane and probed with the anti-crotalic antivenoms manufactured in Instituto Butantan (Panel A) and
Instituto Vital Brazil (Panel B). The two-headed arrow indicates the position of crotamine.

Program, Fundação Carlos Chagas Filho de Amparo à


Acknowledgements Pesquisa do Estado do Rio de Janeiro (FAPERJ), Financiadora
de Estudos e Projetos (FINEP), and Fundação de Amparo a
This work has been financed by grants BFU2007-61563 and Pesquisa do Estado de Minas Gerais (FAPEMIG). CCN and RSR
BFU2010-17373 from the Ministerio de Ciencia e Innovación, are recipients of fellowships from CNPq and FAPEMIG,
Madrid, Spain, 2008/11688-5 and 2009/09698-5 from Fundação respectively. MMSS gratefully acknowledges Fundación Car-
de Amparo a Pesquisa do Estado de São Paulo (FAPESP), olina (http://www.fundacioncarolina.es) for providing a Fel-
and grants from Conselho Nacional de Desenvolvimento lowship of the Mobility Programme for Brazilian University
Científico e Tecnológico (CNPq), CNPq National Institute Professors.
J O U R NA L OF PR O TE O MI CS 7 3 ( 2 01 0 ) 1 7 5 8– 1 7 7 6 1773

Fig. 9 – Immunodepletion of proteins from C. d. terrificus venom by an experimental antivenom generated in rabbits against
C. d. terrificus venom. Reverse-phase separation of the proteins from the crotamine-positive venom of adult C. d. terrificus
(Southern population) recovered after antivenomic treatment of the crude venom with an experimental antivenom raised in
rabbits against the same venom. Insets, left panel, specificity control: reverse-phase separation of the proteins from the
crotamine-positive venom of adult C. d. terrificus (Southern population) recovered after treatment of the crude venom with
non-immune IgG; Right panel: lane a, SDS-PAGE of the C. d. terrificus venom used as immunogen; lane b, molecular mass
markers; lane c, Ponceau red-stained nitrocellulose membrane showing electroblotted C. d. terrificus venom proteins;
lanes d and e, nitrocellulose membranes probed with purified non-immune and immune IgG, respectively. Ctx, crotoxin; Cro,
crotamine.

REFERENCES Sistrurus. Venomous animals and their venoms, vol. 2.


Venomous vertebratesNew York & London: Academic Press;
1971. p. 115–56.
[1] Gloyd HK. The rattlesnakes, Genera Sistrurus and Crotalus: A [10] Campbell JA, Lamar WW. The venomous reptiles of the
study in zoogeography and evolution. Chicago: Chicago Western Hemisphere. Ithaca (NY) and London: Comstock
Academy of Sciences; 1940. p. 1–270. Special Publication Publishing Associates; 2004.
No. 4. [11] Hoser RT. A reclassification of the rattlesnakes; species
[2] Brattstrom BH. Evolution of the pitvipers. Transactions of the formerly exclusively referred to the genera Crotalus and
San Diego Society of Natural History, vol. 13; 1964. p. 185–268. Sistrurus. Aust J Herpetol 2009;6:1–21.
[3] Knight A, Styler D, Pelikan S, Campbell JA, Densmore LD, [12] Wüster W, Ferguson JE, Quijada-Mascareñas JA, Pook CE,
Mindrell DP. Choosing among hypotheses of rattlesnake Salomão MG, Thorpe RS. Tracing an invasion: landbridges,
phylogeny: a best-fit rate test for DNA sequence data. Syst refugia and the phylogeography of the Neotropical
Biol 1993;42:356–67. rattlesnake (Serpentes: Viperidae: Crotalus durissus). Mol Ecol
[4] Klauber LM. Rattlesnakes: their habitats, life histories, and 2005;14:1095–108.
influence on mankind. Second edition. Berkeley: University [13] Quijada-Mascareñas JA, Ferguson JE, Pook CE, Salomão MG,
of California Press; 1997. Thorpe RS, Wüster W. Phylogeographic patterns of
[5] Place AJ, Abramson CI. A quantitative analysis of the trans-Amazonian vicariants and Amazonian biogeography:
ancestral area of rattlesnakes. J Herpetol 2004;38:152–6. the Neotropical rattlesnake (Crotalus durissus complex) as an
[6] Castoe TA, Daza JM, Smith EN, Sasa M, Kuch U, Campbell JA, example. J Biogeogr 2007;34:1296–312.
et al. Comparative phylogeography of pitvipers suggests a [14] Cardoso JLC. Animais peçonhentos no Brasil: biologia clínica
consensus of ancient Middle American highland biogeogra- e terapêutica dos acidentes. São Paulo: Sarvier; 2003.
phy. J Biogeogr 2009;36:88–103. [15] Santoro ML, Sousa-e-Silva MCC, Gonçalves LRC,
[7] Greene H. Snakes: the evolution of mystery in nature. Almeida-Santos SM, Cardoso DF, Laporta-Ferreiro LI, et al.
Berkeley: Univ. of California Press; 1997. Comparison of the biological activities in venoms from three
[8] Parkinson CL, Campbell JA, Chippindale P. Multigene subspecies of South-American rattlesnake Crotalus durissus
phylogenetic analysis of pitvipers, with comments on their terrificus, C. durissus cascavella and C. durissus collilineatus.
biogeography. In: Höggren M, Schuett GW, Greene H, Douglas Comp Biochem Physiol 1999;122C:61–73.
ME, editors. Biology of the vipers. Eagle Mountain, UT: Eagle [16] Oshima-Franco Y, Hyslop S, Prado JF, Cruz-Hoffling MA,
Mountain Publishing; 2002. p. 93–110. Rodriguez LS. Neutralizing capacity of antisera raised in
[9] Klauber LM. Classification, distribution and biology of the horses and rabbits against Crotalus durissus terrificus
venomous snakes of northern Mexico, the United States and (South-American rattlesnake) venom and its main toxin,
Canada. In: Bücherl W, Buckley EE, editors. Crotalus and crotoxin. Toxicon 1999;37:1341–57.
1774 J O U R NA L OF PR O TE O MI CS 73 ( 20 1 0 ) 1 7 5 8– 1 7 7 6

[17] Vital-Brazil O. Pharmacology of crystalline crotoxin. II. proteomic tools in the design and control of antivenoms for
Neuromuscular blocking action. Mem Inst Butantan 1966;33: the treatment of snakebite envenoming. J Proteomics
981–92. 2009;72:165–82.
[18] Azevedo-Marques MM, Hering SE, Cupo P. Acidente crotálico. [37] Chippaux JP, Willians V, White J. Snake venom variability:
In: Cardoso JLC, Franca FOS, Wen FH, Málaque CMS, Haddad methods of study, results and interpretation. Toxicon
V, editors. Animais Peçonhentos no Brasil. Biología, Clínica e 1991;29:1279–303.
Terapeutica dos Acidentes, 2nd editionSao Paulo: Sarvier; [38] Alape-Girón A, Sanz L, Escolano J, Flores-Díaz M, Madrigal M,
2009. p. 108–15. Sasa M, et al. Snake venomics of the lancehead pitviper
[19] Warrell DA. Snakebites in Central and South America: Bothrops asper: geographic, individual, and ontogenetic
epidemiology, clinical features, and clinical management. variations. J Proteome Res 2008;7:3556–71.
In: Campbell JA, Lamar WW, editors. The venomous reptiles [39] Núñez V, Cid P, Sanz L, De La Torre P, Angulo Y, Lomonte B,
of the Western Hemisphere. Ithaca and London: Comstock et al. Snake venomics and antivenomics of Bothrops atrox
Publishing Associates; 2004. p. 709–61. venoms from Colombia and the Amazon regions of Brazil,
[20] Bon C, Bouchier C, Choumet V, Faure G, Jiang MS, Lambezat Perú and Ecuador suggest the occurrence of geographic
MP, et al. Crotoxin, half-century of investigations on a variation of venom phenotype by a trend towards
phospholipase A2 neurotoxin. Act Physiol Pharmacol paedomorphism. J Proteomics 2009;73:57–78.
Latinoam 1989;39:439–48. [40] Warrell DA. Geographical and intraspecies variation in the
[21] Bon C. Multicomponent neurotoxic phospholipases A2. In: clinical manifestations of envenoming by snakes. In: Thorpe
Kini RM, editor. Venom phospholipase A2 enzymes: RS, Wüster W, Malhotra A, editors. Venomous snakes:
structure, function and mechanism. Chichester: Wiley; 1997. ecology, evolution and snakebite. Oxford: Clarendon Press;
p. 269–85. 1997. p. 189–203.
[22] Sampaio SC, Hyslop S, Fontes MR, Prado-Franceschi J, [41] Chippaux JP. Impact of the environment on envenomation
Zambelli VO, Magro AJ, et al. Crotoxin: novel activities for a incidence and severity. Med Sci 2009;25:858–62.
classic beta-neurotoxin. Toxicon 2010;55:1045–60. [42] Fasman DG. Practical Handbook of Biochemistry and
[23] Boldrini-França J, Rodrigues RS, Fonseca FP, Menaldo DL, Molecular Biology. Boston: CRC Press; 1992.
Ferreira FB, Henrique-Silva F, et al. Crotalus durissus [43] Calvete JJ, Juárez P, Sanz L. Snake venomics. Strategy and
collilineatus venom gland transcriptome: analysis of gene applications. J Mass Spectrom 2007;42:1405–14.
expression profile. Biochimie 2009;91:586–95. [44] Altschul SF, Madden TL, Schaffer AA, Zhang J, Zhang Z,
[24] Calvete JJ, Sanz L, Cid P, de la Torre P, Flores-Díaz M, dos Miller W, et al. Gapped BLAST and PSI-BLAST: a new
Santos MC, et al. Snake venomics of the Central American generation of protein database search programs. Nucleic
rattlesnake Crotalus simus and the South American Crotalus Acids Res 1997;25:3389–402.
durissus complex points to neurotoxicity as an adaptive [45] Towbin H, Staehelin T, Gordon J. Electrophoretic transfer of
paedomorphic trend along Crotalus dispersal in South proteins from polyacrylamide gels to nitrocellulose sheets:
America. J Proteome Res 2010;9:528–44. procedure and some applications. Proc Natl Acad Sci USA
[25] Chang CC, Tseng H. Effect of crotamine, a toxin of South 1979;76:4350–4.
American rattlesnake venom, on the sodium channel of [46] Lomonte B, Escolano J, Fernández J, Sanz L, Angulo Y,
murine skeletal muscle. Br J Pharmacol 1978;63:551–9. Gutiérrez JM, et al. Snake venomics and antivenomics of the
[26] Oguiura N, Boni-Mitak M, Rádis-Baptista G. New view arboreal neotopical pitvipers Bothriechis lateralis and
on crotamine, a small basic polypeptide myotoxin from Bothriehis schlegelii. J Proteome Res 2008;7:2445–57.
South American rattlesnake venom. Toxicon 2005;46(18): [47] Gutiérrez JM, Sanz L, Escolano J, Fernández J, Lomonte B,
363–70. Angulo Y, et al. Snake venomic of the Lesser Antillean pit
[27] Lewontin RC. The genetic basis of evolutionary change. vipers Bothrops caribbaeus and Bothrops lanceolatus:
New York: Columbia University Press; 1974. correlation with toxicological activities and
[28] Orr HA, Coyne JA. The genetics of adaptation — a immunoreactivity of a heterologous antivenom. J Proteome
reassessment. Am Nat 1992;140:725–42. Res 2008;7:4396–408.
[29] Golding GB, Dean AM. The structural basis of molecular [48] Calvete JJ, Borges A, Segura A, Flores-Díaz M, Alape-Girón A,
adaptation. Mol Biol Evol 1998;15:355–69. Gutiérrez JM, et al. Snake venomics and antivenomics of
[30] Gibbs HL, Mackessy SP. Functional basis of a molecular Bothrops colombiensis, a medically important pitviper of the
adaptation: prey-specific toxic effects of venom from Bothrops atrox-asper complex endemic to Venezuela:
Sistrurus rattlesnakes. Toxicon 2009;53:672–9. contributing to its taxonomy and snakebite management.
[31] Gibbs HL, Sanz L, Calvete JJ. Snake population venomics: J Proteomics 2009;72:227–40.
proteomics-based analyses of individual variation reveals [49] Gutiérrez JM, Sanz L, Flores-Díaz M, Figueroa L, Madrigal M,
gene regulation effects on venom protein expression in Herrera M, et al. Impact of regional variation in Bothrops
Sistrurus rattlesnakes. J Mol Evol 2009;68:113–25. asper snake venom on the design of antivenoms: integrating
[32] Greene HW. Dietary correlates of the origin and radiation of antivenomics and neutralization approaches. J Proteome Res
snakes. Am Zool 1983;23:431–41. 2010;9:564–77.
[33] Espino-Solis GP, Riaño-Umbarila L, Becerril B, Possani LD. [50] Calvete JJ, Cid P, Sanz L, Segura A, Villalta M, León G, Harrison
Antidotes against venomous animals: state of the art and R, Durfa N, Nasidi A, Theakston RG, Warrell D, Gutiérrez JM.
prospectives. J Proteomics 2009;72:183–99. Antivenomic Assessment of the Immunological Reactivity of
[34] Gutiérrez JM, León G. Snake antivenoms. In: De Lima ME, EchiTAb-Plus-ICP®, an Antivenom for the Treatment of
Pimenta AMC, Martin-Euclaire MF, Zingali RB, editors. Snakebite Envenoming in sub-Saharan Africa Am. J Trop
Animal toxins: state of the art. Perspectives in health and Med Hyg 2010;82:1194–201.
biotechnologyBelo Horizonte, Brazil: Editora UFMG; 2009. [51] Georgieva G, Öhler M, Seifert J, von Bergen M, Arni RK, Genov
p. 393–421. N, Betzel C. Snake venomic of Crotalus durissus terrificuss:
[35] Calvete JJ, Sanz L, Angulo Y, Lomonte B, Gutiérrez JM. correlation with pharmacological activities. J Proteome Res
Venoms, venomics, antivenomics. FEBS Lett 2009;583: 2010;9:2302–16.
1736–43. [52] Sanz L, Escolano J, Ferretti M, Biscoglio MJ, Rivera E, Crescenti
[36] Gutiérrez JM, Lomonte B, León G, Alape-Girón A, Flores-Díaz EJ, et al. Snake venomics of the South and Central American
M, Sanz L, et al. Snake venomics and antivenomics: Bushmasters. Comparison of the toxin composition of
J O U R NA L OF PR O TE O MI CS 7 3 ( 2 01 0 ) 1 7 5 8– 1 7 7 6 1775

Lachesis muta gathered from proteomic versus Cardwell MD, Bush SP, editors. The biology of rattlesnakes.
transcriptomic analysis. J Proteomics 2008;71:46–60. Loma Linda, California: Loma Linda University Press; 2008.
[53] Calvete JJ, Marcinkiewicz C, Sanz L. Snake venomics of Bitis p. 495–510.
gabonica gabonica. Protein family composition, subunit [70] Mackessy SP. Evolutionary trends in venom composition in
organization of venom toxins, and characterization of the Western Rattlesnakes (Crotalus viridis sensu lato):
dimeric disintegrins bitisgabonin-1 and bitisgabonin-2. Toxicity vs. tenderizers. Toxicon 2010;55:1463–74.
J Proteome Res 2007;6:326–36. [71] Furtado MFD, Santos MC, Kamiguti AS. Age-related
[54] Wagstaff SC, Sanz L, Juárez P, Harrison RA, Calvete JJ. biological activity of South American rattlesnake (Crotalus
Combined snake venomics and venom gland transcriptomic durissus terrificus) venom. J Venom Anim Toxins Incl Trop Dis
analysis of the ocellated carpet viper, Echis ocellatus. 2003;9:186–201.
J Proteomics 2009;71:609–23. [72] Powell RL, Lieb CS. Perspectives on venom evolution in
[55] Howgood BJ. Crotoxin, the phospholipase A2 neurotoxin Crotalus. In: Hayes WK, Beaman KR, Cardwell MD, Bush SP,
from the venom of Crotalus durissus terrificus. Mem Inst editors. The biology of rattlesnakes. Loma Linda, California:
Butantan 1990;52:21–2. Loma Linda University Press; 2008. p. 551–6.
[56] Martins AMC, Toyama MH, Havt A, Novello JC, Marangoni S, [73] John TR, Smith LA, Kaiser II. Genomic sequences encoding
Fonteles MC, et al. Determination of Crotalus durissus the acidic and basic subunits of Mojave toxin: unusually high
cascavella venom components that induce renal toxicity in sequence identity of non-coding regions. Gene 1994;139:
isolated rat kidneys. Toxicon 2002;40:1165–71. 229–34.
[57] Amora DN, Souza TS, Martins AMC, Barbosa PSF, Magalhães [74] Tchorbanov B, Grishin E, Aleksiev B, Ovchinnikov Y. A
MR, Toyama MH, et al. Effects of Crotalus durissus collilineatus neurotoxic complex from the venom of the Bulgarian viper
venom in the isolated rat kidney. Toxicon 2006;47:260–4. (Vipera ammodytes meridionalis) and a partial amino acid
[58] Gutiérrez JM, Cerdas L. Mechanism of action of myotoxins sequence of the toxic phospholipase A2. Toxicon 1978;16:
isolated from snake venoms. Rev Biol Trop 1984;32:213–22. 37–44.
[59] De Sousa-e-Silva MC, Tomy SC, Tavares FL, Navajas L, [75] Banumathi S, Rajashankar KR, Nötzel C, Aleksiev B, Singh TP,
Larsson MH, Lucas SR, et al. Hematological, hemostatic and Genov N, et al. Structure of the neurotoxic complex vipoxin
clinical chemistry disturbances induced by Crotalus durissus at 1.4 Å resolution. Acta Crystrallogr 2001;D57:1552–9.
terrificus snake venom in dogs. Hum Exp Toxicol 2003;22: [76] de Haro L, Robbe-Vincent A, Saliou B, Valli M, Bon C,
491–500. Choumet V. Unusual neurotoxic envenomations by Vipera
[60] de Oliveira DGL, Murakami MT, Cintra ACO, Franco JJ, aspis aspis snakes in France. Hum Exp Toxicol 2002;21:137–45.
Sampaio SV, Arni RK. Functional and structural analysis of [77] Ferquel E, de Haro L, Jan V, Guillemin I, Jourdain S, Teynié A,
two fibrinogen-activating enzymes isolated from the d'Alayer J, Choumet V. Reappraisal of Vipera aspis venom
venoms of Crotalus durissus terrificus and Crotalus durissus neurotoxicity. PLoS ONE 2007;2(11):e1194.
collilineatus. Acta Biochim Biophys Sin 2009;41:21–9. [78] Fadel V, Bettendorff P, Hermann T, de Azevedo Jr WF,
[61] Alexander G, Grothusen J, Zepeda H, Schwartzman RJ. Oliveira EB, Yamana T, Wüthrich K. Automated NMR
Gyroxin, a toxin from the venom of Crotalus durissus terrificus, structure determination and disulfide bond identification of
is a thrombin-like enzyme. Toxicon 1988;26:953–60. the myotoxin crotamine from Crotalus durissus terrificus.
[62] Guarnieri MCI, Melo ESLI, Melo KMSII, Albuquerque-Modesto Toxicon 2005;46:759–67.
JCIII, Prieto-da-Silva ARBIV, Rádis-Baptista G. Cloning of a [79] Schenberg S. Geographical pattern of crotamine distribution
novel acidic phospholipase A2 from the venom gland of in the same rattlesnake subspecies. Science 1959;129:1361–3.
Crotalus durissus cascavella (Brazilian northeastern rattle- [80] Gonçalves JM, Arantes EG. Estudos sobre venenos de
snake). J Venom Anim Toxins Incl Trop Dis 2009;15:4. doi: serpentes brasileiras. III — Determinação quantitativa de
10.1590/S1678-91992009000400012. crotamina no veneno de cascavel brasileira. An Acad Bras
[63] Saravia P, Rojas E, Arce V, Guevara C, López JC, Chaves E, Ciênc 1956;28:369–71.
et al. Geographic and ontogenic variability in the venom of [81] Barrio A, Vital Brazil O. Neuromuscular action of the Crotalus
the neotropical rattlesnake Crotalus durissus: terrificus terrificus (Laur.) poisons. Acta Physiol Latinoam
pathophysiological and therapeutic implications. Rev Biol 1951;1:291–308.
Trop 2002;50:337–46. [82] Warren WC, Hillier LW, Marshall Graves JA, Birney E, Ponting
[64] Tavares NA, Correia JM, Guarneri MC, Lima-Filho JL, CP, Grützner F, et al. Genome analysis of the platypus reveals
Prieto-da-Silva AR, Radis-Baptista G. Expression of mRNAs unique signatures of evolution. Nature 2008;453:175–83.
coding for VAP1/crotastatin-like metalloproteases in the [83] Rádis-Baptista G, Kubo T, Oguiura N, Da Silva ARBP, Hayashi
venom glands of three South American pit vipers assessed MAF, Oliveira EB, et al. Identification of crotasin, a
by quantitative real-time PCR. Toxicon 2008;52:897–907. crotamine-related gene of Crotalus durissus terrificus. Toxicon
[65] Toyama MH, Toyama Dde O, Passero LF, Laurenti MD, 2004;43:751–9.
Corbett CE, Tomokane TY, et al. Isolation of a new L-amino [84] Rádis-Baptista G, Kubo T, Oguiura N, Svartman M, Almeida
acid oxidase from Crotalus durissus cascavella venom. Toxicon TMB, Batistic RF, et al. Structure and chromosomal
2006;47:47–57. localization of the gene for crotamine, a toxin from the South
[66] Prado-Franceschi J, Brazil OV. Convulxin, a new toxin from American rattlesnake, Crotalus durissus terrificus. Toxicon
the venom of the South American rattlesnake Crotalus 2003;42:747–52.
durissus terrificus. Toxicon 1981;19:875–87. [85] Oguiura N, Collares MA, Furtado MFD, Ferrarezzi H, Suzuki H.
[67] Murakami MT, Zela SP, Gava LM, Michelan-Duarte S, Cintra Intraspecific variation of the crotamine and crotasin genes
ACO, Arni RK. Crystal structure of the platelet activator in Crotalus durissus rattlesnakes. Gene 2009;446:35–40.
convulxin, a disulfide-linked 44 cyclic tetramer from the [86] Oguiura N, Boni-Mitake M, Rádis-Baptista G. New view on
venom of Crotalus durissus terrificus. Biochem Biophys Res crotamine, a small basic polypeptide myotoxin from South
Commun 2003;310:478–82. American rattlesnake venom. Toxicon 2005;46:363–70.
[68] Batuwangala T, Leduc M, Gibbins JM, Bon C, Jones EY. [87] Vanzolini PE, Ramos-Costa AMM, Vitt LJ. Répteis das
Structure of the snake venom toxin convulxin. Acta Caatingas. Rio de Janeiro: Academia Brasileira de Ciências;
Crystallogr 2004;D60:46–53. 1980. p. 67–8.
[69] Mackessy SP. Venom composition in rattlesnakes: trends [88] Salomão MG, Almeida-Santos SM, Puorto G. Activity pattern
and biological significance. In: Hayes WK, Beaman KR, of Crotalus durissus (Viperidae, Crotalinae): feeding,
1776 J O U R NA L OF PR O TE O MI CS 73 ( 20 1 0 ) 1 7 5 8– 1 7 7 6

reproduction and snake bite. Stud Neotrop Fauna Environ [96] Rodriguez JP, De Marzi M, Maruñak S, Malchiodi EL, Leiva LC,
1995;30:101–6. Acosta O. Rabbit IgG antibodies against phospholipase A2
[89] Mancin AC, Soares AM, Adrião-Escarso SH, Faça VM, Greene from Crotalus durissus terrificus neutralize the lethal activity
LJ, Zuccolotto S, et al. The analgesic activity of crotamine, a of the venom. Medicina (B Aires) 2006;66:512–6.
neurotoxin from Crotalus durissus terrificus (South American [97] Beghini DC, Hernandez-Oliveira S, Rodrigues-Simioni L,
rattlesnake) venom: a biochemical and pharmacological Novello JC, Hyslop S, Marangoni S. Anti-sera raised in rabbits
study. Toxicon 1998;36:1927–37. against crotoxin and phospholipase A2 from Crotalus durissus
[90] Fry BG, Wickramaratna JC, Jones A, Alewood PF, Hodgson cascavella venom neutralize the neurotoxicity of the venom
WC. Species and regional variations in the effectiveness of and crotoxin. Toxicon 2004;22:141–8.
antivenom against the in vitro neurotoxicity of death adder [98] Sano-Martins IS, Tomy SC, Campolina D, Dias MB, de Castro
(Acanthophis) venoms. Toxicol Appl Pharmacol 2001;175:140–8. SC, de Sousa-e-Silva MC, et al. Coagulopathy following lethal
[91] Judge RK, Henry PJ, Mirtschin P, Jelinek G, Wilce JA. Toxins and non-lethal envenoming of humans by the South
not neutralized by brown snake antivenom. Toxicol Appl American rattlesnake (Crotalus durissus) in Brazil. QJM
Pharmacol 2006;213:117–25. 2001;94:551–9.
[92] Correa-Netto C, Teixeira-Araujo R, Aguiar AS, Melgarejo AR, [99] Bourillet F. Comparative neuromuscular action of crotamine
De-Simone SG, Soares MR, et al. Immunome and venome of and veratrine. Ann Pharm Fr 1970;28:535–44.
Bothrops jararacussu: a proteomic approach to study the [100] Cheymol J, Gonçalves JM, Bourillet F, Roch-Arveiller M. A
molecular immunology of snake toxins. Toxicon 2010;55: comparison of the neuromuscular action of crotamine and
1222–35. the venom of Crotalus durissus terrificus var. crotaminicus.
[93] De Oliveira RC, Wen FH, Sifuentes DN. Epidemiologia dos Isolated preparations. Toxicon 1971;9:287–9.
accidentes poe animais peçonhentos. In: Cardoso JLC, França [101] Hayashi MA, Nascimento FD, Kerkis A, Oliveira V, Oliveira
FOS, Wen FH, Málaque CMS, Haddad V, editors. Animais EB, Pereira A, et al. Cytotoxic effects of crotamine are
Peçonhentos no Brasil. Biologia, Clínica e Terapêutica dos mediated through lysosomal membrane permeabilization.
Acidentes, 2nd editionSao Paulo: Sarvier; 2009. p. 6–21. Toxicon 2008;52:508–17.
[94] Jorge MT, Ribeiro LA. Epidemiology and clinical picture of [102] Gutiérrez JM, Dos Santos MC, Furtado MF, Rojas G.
accidental bites by the South American rattlesnake (Crotalus Biochemical and pharmacological similarities between the
durissus). Rev Inst Med Trop São Paulo 1992;34:347–54. venoms of newborn Crotalus durissus durissus and adult
[95] Brazilian Pharmacopeia. Brazilian Pharmacopeia Crotalus durissus terrificus rattlesnakes. Toxicon 1991;29:
Convention. 4nd ed. São Paulo: Atheneu; 2002. 1273–7.

View publication stats

You might also like