You are on page 1of 9

Toxin Reviews

ISSN: 1556-9543 (Print) 1556-9551 (Online) Journal homepage: https://www.tandfonline.com/loi/itxr20

Low molecular mass natural and synthetic


inhibitors of snake venom metalloproteinases

Lina María Preciado & Jaime Andrés Pereañez

To cite this article: Lina María Preciado & Jaime Andrés Pereañez (2018) Low molecular mass
natural and synthetic inhibitors of snake venom metalloproteinases, Toxin Reviews, 37:1, 19-26,
DOI: 10.1080/15569543.2017.1309550

To link to this article: https://doi.org/10.1080/15569543.2017.1309550

Published online: 04 Apr 2017.

Submit your article to this journal

Article views: 99

View related articles

View Crossmark data

Citing articles: 2 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=itxr20
http://informahealthcare.com/txr
ISSN: 1556-9543 (print), 1556-9551 (electronic)

Toxin Rev, 2018; 37(1): 19–26


ß 2017 Informa UK Ltd, trading as Taylor & Francis Group. DOI: 10.1080/15569543.2017.1309550

REVIEW ARTICLE

Low molecular mass natural and synthetic inhibitors of snake venom


metalloproteinases
Lina Marı́a Preciado and Jaime Andrés Pereañez

Programa de Ofidismo/Escorpionismo, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia, Medellı́n, Colombia

Abstract Keywords
Snake venom metalloproteinases (SVMPs) play an important role in local and systemic Inhibitors, snake venom, metalloproteinases,
manifestations of viperid snakebite envenomations. The therapy available has been based on docking, snake bites, envenomations
the intravenous administration of antivenoms; nevertheless, they have a limited efficacy against
local effects provoked by SVMPs. For this reason, it is important to search for alternative venom History
inhibitors, either synthetic or natural, that would complement the action of antivenoms,
particularly regarding neutralization of local tissue damage caused by SVMPs. In this review, Received 19 January 2017
information about low molecular mass synthetic and natural molecules that have been Revised 17 March 2017
reported to be inhibitors of SVMPs is described, focusing on their ability to abrogate the Accepted 18 March 2017
activities of SVMPs and on the molecular mechanisms involved. Published online 31 March 2017

Introduction edema, myonecrosis, dermonecrosis, disruption of hemostasis


mediated by procoagulant or anticoagulant effects, platelet
Snake venom metalloproteinases (SVMPs) are major compo-
aggregation, apoptosis and pro-inflammatory activities (Dı́az
nents of most viperid and some ‘‘colubrid’’ venoms
et al., 2005; Gâz Florea et al., 2016; Gutiérrez & Rucavado,
(Markland & Swenson, 2013). These enzymes are zinc-
2000; Gutiérrez et al., 2005). The mechanism of action of
dependent metalloproteinases which belong to the family of
hemorrhagic SVMPs has been investigated in various ways,
‘‘metzincins’’, together with astacins, serralysins, matrix
and there is consensus in that hydrolysis of basement
metalloproteinases (MMPs) and ADAMs (enzymes with a
membrane (BM) components in microvessels is a key step
disintegrin and metalloproteinase domains). With few excep-
in this process (Escalante et al., 2011; Herrera et al., 2015).
tions, these proteinases contain a similar zinc-binding motif in
In addition, they are able to hydrolyze endothelial cell
their catalytic domain, characterized by the sequence
membrane proteins, such as integrins and cadherins, involved
HEXXHXXGXXH, followed by a Met-turn (Bode et al.,
in cell–matrix and cell–cell adhesion (Gutiérrez et al., 2005).
1993). They are large multi-domain proteins that are classi-
Moreover, disintegrin-like and cysteine-rich domains contain
fied as P-I, P-II and P-III based on the presence or absence of
exosites that interact with endothelial cell integrins, interfer-
non-catalytic ancillary domains that extend beyond the mature
ing with their adhesion to extracellular matrix (Escalante
proteinase domain. While P-I (20–30 kDa proteins) has only
et al., 2011; Gutiérrez & Rucavado, 2000; Gutiérrez et al.,
a proteinase domain, the P-II SVMPs (30–60 kDa) contain
2005).
proteinase and disintegrin domains (Fox & Serrano, 2008).
Furthermore, the P-III SVMPs (60–100 kDa) contain pro-
teinase, disintegrin-like and cysteine-rich domains.
Inhibitors of snake venom metalloproteinases
Additionally, several subclasses have been described within Local tissue damage, such as hemorrhage, myonecrosis,
each class, depending on whether they are monomers or homo dermonecrosis and edema, constitutes one of the most
or heterodimers, and also on the variable patterns of post- common and serious consequences of Viperidae snakebite
translational cleavage of several domains (Fox & Serrano, envenomation. The pathogenesis of these alterations is
2010; Moura-da-Silva et al., 2016). induced by different toxins present in the venoms, such as
A diverse spectrum of biochemical and biological myotoxic phospholipases A2 and hemorrhagic metalloprotei-
activities has been described for the SVMPs, showing nases (Gutiérrez et al., 1998). The therapy for snakebite
therefore their critical role in the overall toxicity of the envenomations has been based on the intravenous adminis-
venom, and their functional versatility. These enzymes are tration of horse and sheep-derived antivenoms (Gutiérrez
responsible for the local and systemic hemorrhagic activity, et al., 2003). However, it has been demonstrated that
neutralization of local effects by antivenoms is difficult
to achieve, not only due to the variety of components
Address for correspondence: Jaime Andrés Pereañez, Facultad de
Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia, inducing these alterations, but also to the rapid onset of
Calle 70 52-21, Medellı́n, Colombia. E-mail: andrespj20@gmail.com these pathological changes (Gutiérrez & Lomonte, 1989).
20 L. M. Preciado & J. A. Pereañez Toxin Rev, 2018; 37(1): 19–26

Thus, it is important to search for alternative venom the pathological study of thigh muscles of the ddY strain
inhibitors, either synthetic or natural, that would complement white mice showed that RA inhibited hemorrhage and
the action of antivenoms, particularly regarding neutralization neutrophil infiltrations (Aung et al., 2010).
of local tissue damage (Pereañez et al., 2013). In this work, On the other hand, Srinivasa et al. (2014) reported the
the main types of low molecular mass molecules that have inhibitory effect of compound 5d (Figure 1B), an apigenin
been shown to have inhibitory potential on SVMPs will be based molecule, against SVMPs effects in the venom of Echis
summarized. However, natural inhibitors of SVMPs of carinatus (EC), both in silico and in vivo. This molecule
proteinaceous nature were not included, since they have effectively abrogated EC venom-induced local hemorrhage,
been reviewed in a recent work (Bastos et al., 2016). tissue necrosis and myotoxicity in a dose-dependent fashion.
Nevertheless, this information is of value as a starting point The histopathological study showed effective inhibition of
for future research in this field. BM degradation and accumulation of inflammatory leuco-
cytes at the site of venom injection. The compound also
inhibited venom-induced fibrin and fibrinogen degradation.
Phenolic compounds
Molecular docking studies of compound 5d and bothropasin,
Many current studies focus on the health benefits of a P-III metalloproteinase from the venom of Bothrops
phytochemicals, especially their antioxidant and antimicrobial jararaca, showed that the flavone group of compound 5d
properties (Gyawali & Ibrahim, 2014; Soobrattee et al., 2005). was found buried into the active site grove via strong
It is known that these compounds exert preventive activity hydrogen bonding between the hydroxyl group and Glu146 of
against infectious, degenerative diseases, inflammation and hydrophobic pocket that connects with Ile142 and His145 and
allergies via antioxidant antimicrobial and proteins/enzymes other hydrophobic key amino acids, like Val141, Ile168,
neutralization/modulation mechanisms (Ozcan et al., 2014). Gly170, Pro171, Thr172 and Ile173. Additionally, acyl
The ability of some phenolic compounds to inhibit SVMPs bromide tail of compound 5d formed hydrogen bonding
has been tested; as an example, it has been established that with residues of active site including Thr110, Ile111 and
rosmarinic acid (RA) (Figure 1A) inhibits hemorrhagic Gly112 (Srinivasa et al., 2014).
activity induced by the whole venoms of Trimeresurus Other studies with phenolic compounds have been carried
flavoviridis, Crotalus atrox and Gloydius blomhoffii, or by out with gallic acid (Figure 1C) against the local toxicity of
purified toxins, like HTb isolated from the venom of C. atrox, Daboia russelli venom and its purified hemorrhagic complex.
bilitoxin-2 from Agkistrodon bilineatus, Ac1-proteinase from Gallic acid inhibited proteolytic activity in vitro and hemor-
Deinagkistrodon acutus and HT-1 from Bitis arietans venom. rhage, edema-forming and dermo- and myonecrotic activities
This compound also prevents degradation of human fibrino- of both D. ruselli venom and hemorrhagic complex in in vivo
gen and abrogates type IV collagen hydrolysis by incubation experiments. In this study, it was demonstrated that the
with the venom in a concentration of 0.5 mg/mL. Moreover, inhibition was due to gallic acid induced structural changes in

Figure 1. Phenolic inhibitors. (A) Rosmarinic acid. (B) 5d compound, apigenin derivate. (C) Gallic acid. (D) Isoquercitrin. (E) Myricetin-3-O-
glucoside. (F) Gallocatechin.
DOI: 10.1080/15569543.2017.1309550 Inhibitors of snake venom metalloproteinases 21

hemorrhagic complex as revealed by alterations in fluores- inhibitors might lead to new therapies for acute and chronic
cence emission and Circular Dichroism Spectral Studies inflammatory and vascular diseases (Hu et al., 2007). MMPs
(Mahadeswaraswamy et al., 2011). are grouped together with astacins, serralysins and reproly-
Moreover, medicinal plant extracts are rich sources of sins, including SVMPs and ADAMs, within the superfamily
natural inhibitors and have been shown to antagonize the of ‘‘metzincins’’, characterized by similar structural features
activity of some venoms and metalloproteinases. As an at the catalytic site and a neighboring loop (Bode et al., 1993).
example, isoquercitrin (Figure 1D), myricetin-3-O-glucoside The high degree of structural and functional homology
(Figure 1E) and gallocatechin (Figure 1F), isolated from between SVMPs and their mammalian relatives, MMPs,
lyophilized aqueous extract of Schizolobium parahyba leaves, suggests that substrate/inhibitor interactions between these
neutralized the biological and enzymatic activities of subfamilies can be similar (Howes et al., 2007). Most MMP
Bothrops venoms and the isolated metalloproteinase inhibitors developed to date consist of a zinc-binding group
BjussuMP-I, from B. jararacussu venom. The flavonol (ZBG), which binds the catalytic metal ion, and a peptido-
glycoside isoquercitrin was able to inhibit hemorrhage mimetic backbone that interacts noncovalently with the active
induced by the venom, but did not show a high efficiency site of the enzyme. Nevertheless, due to the presence the ZBG
to inhibit fibrinogenolytic activity. On the contrary, myricetin- in these molecules, a big part of their activity can be
3-O-glucoside showed a potent inhibition of the fibrinogen explained for their capacity to chelate zinc. Several of these
proteolytic activity of B. alternatus venom but it did not synthetic inhibitors have been shown to effectively neutralize
inhibit hemorrhage, which may indicate a specific action enzymatic and toxic activities of SVMPs (Escalante et al.,
against serine proteinase enzymes. On the other hand, 2000; Rucavado et al., 2004).
gallocatechin was efficient in neutralizing the hemorrhagic The effectiveness of matrix metalloproteinases inhibitors
and fibrinogenolytic activity induced by B. alternatus, B. (MMPIs) such as Marimastat (Figure 2A), AG-3340
jararacussu venom and BjussuMP-I metalloproteinase (Vale (Prinomastat) (Figure 2B), Bay-12 9566 (Tanomastat)
et al., 2011). (Figure 2C) and CGS-270 23A (Figure 2D) in abrogating
the hemorrhagic and proteolytic activities of whole
E. ocellatus venom and EoVMP2 (a hemorrhagic P-III
Matrix metalloproteinase inhibitors
metalloproteinase) was demonstrated (Howes et al., 2007).
Matrix metalloproteinases have outgrown the field of extra- The MMPIs Marimastat and CGS-270 23A are both
cellular matrix biology and have progressed toward being hydroxamic acid derivatives that are designed to exert
important regulatory molecules in cancer and inflammation. broad-range MMP inhibition by mimicking the cleavage site
This rise in status was accompanied by the development of of MMP (collagen) substrate. On the other hand, AG-3340
various types of inhibitors aimed at controlling the role of and Bay-12 9566 are both designed to be selective inhibitors
these enzymes in disease. Although clinical trials with of the gelatinases MMP-2 and MMP-9 (Heath & Grochow,
synthetic inhibitors for the treatment of cancer were disap- 2000; Hidalgo & Eckhardt, 2001; Yip et al., 1999).
pointing, recent data indicate that the use of selective These compounds were effective in neutralizing the

Figure 2. MMP inhibitors. (A) Marimastat. (B) Prinomastat. (C) Tanomastat. (D) CGS-270 23A. (E) Batimastat.
22 L. M. Preciado & J. A. Pereañez Toxin Rev, 2018; 37(1): 19–26

hemorrhagic activity of EoVMP2 in murine assays, but only Wat67. Wat67 has been proposed to play a critical role as the
AG-3340 successfully inhibited the hemorrhagic activity of catalytic nucleophile in SVMPs, attacking the scissile peptide
whole E. ocellatus venom. bond after polarization by the highly conserved Glu143
In a similar way, the peptidomimetic Batimastat (BB-94) (Lingott et al., 2009).
(Figure 2E) was assessed to determinate its ability to These results demonstrate that MMPIs are a promising
neutralize the local effects induced by the purified B. asper source of compounds with the ability to inhibit the local and
metalloproteinase BaP1 (Escalante et al., 2000) and the systemic effects of SVMP. Nevertheless, MMPIs show
systemic effects induced by the whole venom (Rucavado specificity toward SVMPs and their application to the
et al., 2004). This compound inhibited proteolytic, hemor- neutralization SVMP effects may require a synthetic MMPI
rhagic, dermonecrotic and edema forming activities of this mixture, ensuring that a close structural component for each
metalloproteinase when incubated with the enzyme prior SVMP is represented (Howes et al., 2007).
to the assays. When the inhibitor was administered i.m. at
the site of the toxin injection without preincubation, rapidly
Zinc chelating agents
after metalloproteinase administration, it totally abrogated
the hemorrhagic and dermonecrotic effects of BaP1. SVMPs are zinc-containing endopeptidases, and are therefore
Nevertheless, inhibition was less effective as the time lapse dependent on the presence of zinc ions at the catalytic site (for
between toxin and batimastat injection increased, due to the their enzymatic activity) and calcium ions in other molecular
extremely rapid development of BaP1-induced local tissue regions (for the structural stability of the enzyme). Chelation
damage (Escalante et al., 2000). In addition, Batimastat of these metal ions by agents such as EDTA (Figure 3A)
inhibited lethality when a venom challenge dose of two LD50s (ethylenediamine-tetraacetic acid) and o-phenanthroline
was used by intraperitoneal and intravenous routes, also (Figure 3B) has shown to inhibit SVMP activity in several
inhibited the hemorrhagic effect induced by the venom in studies (Borkow et al., 1997; Howes et al., 2007; Mazzi et al.,
lungs after intravenous injection, and exerted a significant 2004; Patiño et al., 2010).
inhibition of in vitro coagulant and in vivo defibrinogenating Specific zing chelating agents such as N, N, N0 , N0 -Tetrakis
effects of venom. It is suggested that Batimastat may act (2-pyridylmethyl) ethane-1,2-diamine (TPEN) (Figure 3C),
with similar mechanism of peptidomimetic inhibitor with a diethylene triamine pentaacetic acid (DTPA) (Figure 3D) and
hydroxamate moiety, which was co-crystalized with BaP1 tetraethylthiuram disulfide (TTD) (Figure 3E) have been
(Lingott et al., 2009). One of the most important findings of evaluated in the inhibition of hemorrhage and myotoxicity
this work was that in the non-complexed structure, BaP1 had induced by E. carinatus (ECV) venom. These compounds
a four-fold zinc coordination system, the zinc ion being completely blocked the hemorrhagic and myotoxic activities
tetrahedrally coordinated by the Ne2 atoms of three histidines of ECV in a dose dependent manner upon co-injection;
(His142, His146 and His152) and a catalytic water. In however, only TPEN successfully neutralized hemorrhage and
contrast, in complexed structure the zinc ion was five-fold myotoxicity following independent injection. Histological
coordinated, with nearly perfect square pyramidal coordin- examinations revealed that TPEN effectively prevents deg-
ation geometry. The Ne2 atoms of His146 and His152 and radation of dermis and BM surrounding the blood vessels in
both oxygens of hydroxamate moiety of inhibitor, formed the mouse skin sections and prevents muscle necrosis and
basal plane of the pyramid, and the Ne2 atom of His142 was accumulation of inflammatory cells at the site of ECV
the apex. The zinc ion was slightly displaced toward the apex, injections. Subsequent studies confirmed, by spectral and
out of the plane of the basal atoms. In addition, the binding of docking experiments, that TPEN has high affinity for Zn2+.
the inhibitor resulted in the displacement of several water This specificity explains the potent inhibition of ECV
molecules at the docking site of native BaP1, i.e. the catalytic metalloproteinases (ECVMPs) in vitro (IC50: 6.7 mM)

Figure 3. Zinc chelating agents. (A) EDTA. (B) o-phenanthroline. (C) TPEN. (D) DTPA. (E) TTD. (F) Clodronate. (G) Doxycycline.
DOI: 10.1080/15569543.2017.1309550 Inhibitors of snake venom metalloproteinases 23

(Nanjaraj Urs et al., 2015). However, the major limitation of hemorrhagic, fibrinogenolytic and caseinolytic activities of
TPEN is its toxicity as it may sequester physiologically class P-I and P-III SVMPs isolated from B. neuwiedi and B.
important Zn2+ containing enzymes at higher doses. jararacussu venoms. However, this compound failed to inhibit
Other two groups of molecules that constitute SVMP the coagulant activity and only partially inhibited the edema
inhibitors are biphosphonates and tetracyclines. Several induced by crude venoms (Januário et al., 2004). Molecular
studies have addressed their inhibition mechanism in various dynamics simulations were used to study the binding of
MMPs, and they have shown that these compounds sequester clerodane diterpenoid to the SVMP (Chinnasamy et al.,
the zinc present at the catalytic site of these enzymes, in 2014).
addition to other actions related with MMP expression and In the same way, the activity of triacontyl p-coumarate
activation (Acharya et al., 2004; Teronen et al., 1999). The (PCT) (Figure 4B), isolated from the root bark of
effectiveness of the biphosphonate clodronate (Figure 3F) and Bombacopsis glabra vegetal extract was assayed against
the tetracycline doxycycline (Figure 3G) to inhibit proteo- harmful effects of Bothropoides pauloensis snake venom and
lytic, hemorrhagic, coagulant and defibrinogenating effects of the SVMPs BleucMP and jararhagin from B. leucurus and B.
B. asper venom was tested. Both compounds were able to jararaca venoms, respectively. This compound neutralized
inhibit these activities, at concentrations in the mM range, fibrinogenolytic activity and plasmatic fibrinogen depletion
when incubated with venom prior to testing. However, when induced by B. pauloensis venom and the isolated SVMPs.
inhibition of hemorrhage was assessed in assays involving PCT also inhibited the hemorrhagic and myotoxic activities
independent injection of venom and drug, inhibition was poor, induced by Jararhagin at 1:5 ratio (toxin:inhibitor, w/w) when
even when these compounds were injected immediately after previously incubated with PCT or when PCT was adminis-
envenomation. These findings support the concept that the tered after toxin injection. Docking studies and molecular
effectiveness of compounds, such as clodronate and doxy- dynamic simulations using the structure of a metalloprotei-
cycline, whose inhibitory action on SVMPs is based on zinc nase (Neuwiedase) suggested that the binding between the
chelation alone, is limited, and stress the view that more protein and the inhibitor occurs mainly in the active site
specific molecules are required for an effective inhibition of region causing blockade of the enzymatic reaction by
SVMPs in vivo (Rucavado et al., 2008). displacement of catalytic water molecule (Mendes et al.,
2013).
Furthermore, the antiproteolytic and antihemorrhagic
Terpenes
properties of triterpenoid saponins named macrolobin-A and
A clerodane diterpenoid known as Bt-CD (7alpha-hydroxy- B (Figure 4C) from Pentaclethra macroloba have been
3,13-clerodadiene-16,15:18,19-diolide) (Figure 4A), isolated reported (da Silva et al., 2007). These compounds were able
from the aerial parts of the Brazilian plant Baccharis trimera to significantly inhibit hemorrhagic and proteolytic activities
(Less), has demonstrated full inhibition of the proteolytic and of Bothrops venoms and isolated BjussuMP-I metalloprotease
hemorrhagic activities of several Bothrops venoms. from B. jararacussu venom. Nevertheless, the IC50 of
Additionally, the inhibitor was able to neutralize the macrolobin A is approximately 0.5 mM for the proteolytic

Figure 4. Terpenes informed as SVMPs inhibitors. (A) Bt-CD. (B) Triacontyl p-coumarate. (C) Macrolobin A, R¼CH2OH Macrolobin B, R¼CH3.
(D) Linearol. (E) Isolinearol. (F) Compound ‘‘8’’. A lupeol acetate derivative.
24 L. M. Preciado & J. A. Pereañez Toxin Rev, 2018; 37(1): 19–26

activity and 1.8 mM for hemorrhage inhibition, showing a 2W12), a SVMP of B. asper venom. The theoric binding
lower potency when compared, for example, with Bt-CD energy was –7.24 kJ/mol. The compound showed hydrophobic
(IC50 ¼ 0.3 mM and 1.0 mM for each bioactivity) (Januário interaction with amino acid residues in the active site pocket
et al., 2004) and batimastat (IC50 ¼ 80 nM for the proteolytic region, which comprises 15 residues of the active site of the
activity and 1.95 mM for hemorrhage inhibition) (Escalante enzyme, which are in close proximity to the ligand and are
et al., 2000). hydrophobic in nature. Alteration of the spectral properties of
Moreover, the secodolastane diterpenes linearol (Figure compound 8 by the added Zn2+ further supports its possible
4D) and isolinearol (Figure 4E), isolated from the Brazilian chelating property (Katkar et al., 2015).
marine brown alga, Canistrocarpus cervicornis, were eval-
uated against some of the toxic effects induced by B. jararaca Other synthetic inhibitors
venom. The mixture of diterpenes inhibited proteolysis in a
Several synthetic quinolinones were tested for its inhibitory
concentration-dependent manner and abrogated the hemor-
properties against hemorrhage caused by different Bothrops
rhagic activity of B. jararaca venom, but failed to inhibit
venoms and isolated SVMPs. The molecule 2-hydroxymethyl-
clotting and edematogenic activities, and thus, would not
6-methoxy-1,4-dihydro-4-quinolinone (Q8) (Figure 5A) was
prevent inflammation induced by snake venoms (Domingos
the most effective compound for inhibiting hemorrhagic and/
et al., 2015).
or proteolytic activities of the crude venoms of B.
Although there are few reports of compounds isolated from
jararacussu, B. moojeni and B. alternatus and the isolated
natural products in the inhibition of SVMPs-induced local and
SVMPs BjussuMP-I, from B. jararacussu, and neuwiedase,
systemic effects, given that most of them are limited to
from B. neuwiedi. Docking and molecular dynamic simula-
demonstrate the biological activity of extracts or fractions,
tions demonstrated that Q8 takes part in a regular square
studies whit C. cervicornis, P. macroloba and B. trimera have
pyramidal coordination geometry of the cofactor Zn2+, which
demonstrated that natural products are a promising alternative
is completed by the Ne2 atoms from active site His and the
to finding new and chemically diverse inhibitors of SVMPs.
Glu142/Zn2+-coordinated catalytic water molecule, and pre-
On the other hand, synthetic terpenes derivatives have been
sents a root-mean-square deviation (RMSD) value of only
tested in the inhibition of E. carinatus venom-induced
0.312 Å in relation to the ideal geometry of this type of
hemorrhage. Lupeol, lupeol acetate and synthetic derivatives
coordination (Baraldi et al., 2016). Therefore, the experimen-
were screened for inhibition of EC venom-induced hemor-
tal and theoretical data indicate that quinolinones could be
rhage in a mouse model; compound 8 (Figure 4F) was found
interesting drug-candidate compounds for snakebite
to be the most potent. This compound neutralized EC venom-
envenoming therapy, particularly for the treatment of local
induced hemorrhage, dermonecrosis and myonecrosis. It also
effects such as hemorrhage.
protected PMNs (polymorphonuclear leukocytes), PBMCs On the other hand, the simplest a-hydroxy acid, 2-
(peripheral blood mononuclear cells) and platelets from hydroxyethanoic acid or glycolic acid (Figure 5B) has been
venom-induced oxidative stress, and offered protection tested in the inhibition of enzymatic, hemorrhagic and edema-
against venom-induced proteolytic cleavage of integrin inducing activities of BaP1, a P-I metalloproteinase isolated
a2b1, GP VI, DDR1 and CX3CR1 receptors present in from B. asper venom. Glycolic acid inhibited the proteolytic
these inflammatory cells, hence favoring their firm binding, activity of BaP1 on azocasein with an IC50 of 1.67 mM, and
which is critical and essential for resolving inflammation and also the hemorrhagic activity in skin and muscle in experi-
tissue repair. Docking studies revealed the binding of ments involving preincubation of enzyme and inhibitor prior
compound 8 in the active site pocket of BaP1 (PDB ID: to injection. Moreover, glycolic acid inhibited, in a

Figure 5. Other synthetic compounds described as SVMPs inhibitors. (A) 2-Hydroxymethyl-6-methoxy-1,4-dihydro-4-quinolinone (Q8). (B) Glycolic
acid. (C) N-acetylcysteine. (D) 1-(3-Dimethylaminopropyl)-1-(4-fluorophenyl)-3-oxo-1,3-dihydroisobenzofuran-5-carbonitrile (DFD). (E) N-(Furan-2-
yl)carbonyl-Leu-l-Trp(P)-(OH)2. (F) Phenazopyridine hydrochloride. (G) Nitrofural. (H) Palmatine chloride. (I) Methoxy-6-harmalan.
DOI: 10.1080/15569543.2017.1309550 Inhibitors of snake venom metalloproteinases 25

concentration-dependent manner, edema-forming activity of were carried out (Villalta-Romero et al., 2012). The virtual
BaP1 in the mouse footpad test. Molecular docking studies screening of the 880 compounds in the Chemical Library was
suggested that glycolic acid forms hydrogen bonds with developed by a fluorogenic method. Consequently, phenazo-
residues Glu143, Arg110 and Ala111 of the enzyme. pyridine HCl (5 F) (IC50 54.3 mM), methoxy 6-harmalan (5 G)
Additionally, molecular modeling and UV–vis spectra studies (IC50 70.4 mM), nitrofural (5 H) (IC50 88.1 mM) and palmatine
suggested that the inhibitor chelates Zn2+ and may occupy chloride (5I) (IC50 93.3 mM) were the most active compounds
part of substrate binding cleft of BaP1 (Pereañez et al., 2013). identified. On the other hand, with the rational peptide design,
These results suggest that glycolic acid is a candidate for the the hydroxamate-containing peptides were more efficient
development of metalloproteinase inhibitors, and it is import- inhibitors of BaP1 than the parental peptides, when Phe-Leu-
ant to determinate the activity of wide range of a-hydroxy Phe (FLF) and Phe-Leu-Arg (FLR) sequences were coupled to
acids in order to find out their inhibitory potential. hydroxamate, with IC50 values of 0.5 mM and 4.6 mM,
On the other hand, N-acetyl cysteine (NAC) (Figure 5C) respectively. Molecular docking analysis suggested different
has been shown to inhibit gelatinase, hyaluronidase, hemor- binding modes evidencing backbone flexibility due to the
rhagic and defibrinogenating activities of Vipera russelli and rotatable bonds. In addition, the best ranking solutions
E. carinatus venoms. NAC inhibited these activities dose generated bidentate binding of the hydroxamate group to
dependently, showed complete inhibition of hemorrhagic zinc, and was also defined that distances below 3.0 Å
activity when incubated with venom prior to testing, whereas generates an optimal binding affinity. Summarizing, this
little inhibition was observed when venom and NAC were study demonstrated that the synthesis of peptides with affinity
injected independently. Inhibition of the BM degradation and for the metalloproteinase active site, coupled to ZBGs, offers
accumulation of inflammatory leukocytes at the site of venom promising possibilities. These results should stimulate future
injection in histological sections further corroborated the work based on further structural modifications of the
inhibitory property of NAC. The observed inhibition of molecules that exerted inhibition of this SVMP (Villalta-
hemorrhage was likely due to zinc chelation as supported by Romero et al., 2012).
spectral studies. Further, docking predictions suggested the
role of –SH and –NH–CO–CH3 groups of NAC in the Acknowledgements
inhibition of SVMPs. Future studies related to the protective
role of NAC against hemorrhage induced by purified SVMPs The authors are thankful to Professor José Marı́a Gutiérrez,
and crude venoms are needed for determining its therapeutic Instituto Clodomiro Picado, for his suggestions and for
potential (Sunitha et al., 2011a). improving the text. The authors acknowledge Comité para
In addition, the inhibitory effect of the synthetic compound el desarrollo de la investigación (CODI-CIQF-217) and
1-(3-dimethylaminopropyl)-1-(4-fluorophe-nyl)-3-oxo-1,3- Universidad de Antioquia (UdeA).
dihydroisobenzofuran-5-carbonitrile (DFD) (Figure 5D) on
viper venom-induced hemorrhagic activity has been tested. Declaration of interest
This compound is a derivative of citalopram, a clinically The authors report no conflicts of interest.
approved antidepressant drug. DFD effectively neutralized the
hemorrhagic activity of medically important viper venoms
Geolocation information
such as E. carinatus, E. ocelatus, E. carinatus sochureki, E.
carinatus leakeyi and Crotalus atrox in a dose-dependent Our research is performed in Medellı́n, Colombia
manner with inhibitory concentrations in the mM range. (Geographic coordinates: 6 130 5500 N 75 340 0500 O).
Histological examinations revealed that the compound DFD
effectively neutralized degradation of the BM, and the ORCID
accumulation of inflammatory leucocytes at the site of E.
Jaime Andrés Pereañez http://orcid.org/0000-0001-9612-
carinatus venom injection further confirms the inhibition of
2827
hemorrhagic activity. According to the docking studies, DFD
binds to the hydrophobic pocket of SVMP with the ki of
19.26  109 kcal/mol without chelating Zn2+ in the active
site (Sunitha et al., 2011b). References
Other synthetic compounds, such as phosphonate analogs Acharya MR, Venitz J, Figg WD, Sparreboom A. (2004). Chemically
of the peptidomimetic N-(Furan-2-yl) carbonyl-Leu-Trp-OH, modified tetracyclines as inhibitors of matrix metalloproteinases.
have been tested. N-(Furan-2-yl) carbonyl-Leu-L-Trp(P)- Drug Resist Updat 7:195–208.
Aung HT, Nikai T, Komori Y, et al. (2010). Biological and pathological
(OH)2 (Figure 5E) caused a 75-fold increase of potency of studies of rosmarinic acid as an inhibitor of hemorrhagic Trimeresurus
the inhibitor against adamalysin II, with an IC50 of 0.4 mM. In flavoviridis (habu) Venom. Toxins 2:2478–89.
this study, it was proposed that the phosphonate group binds Baraldi PT, Magro AJ, Matioli FF, et al. (2016). A novel synthetic
quinolinone inhibitor presents proteolytic and hemorrhagic inhibitory
the catalytic zinc ion in an asymmetric bidentate mode (2.0
activities against snake venom metalloproteases. Biochimie 121:
and 2.8 Å Zn–O distances) (D’Alessio et al., 1999). 179–88.
New strategies such as compound screening and rational Bastos VA, Gomes-Neto F, Perales J, et al. (2016). Natural inhibitors of
peptide design have been tested with the aim of developing snake venom metalloendopeptidases: history and current challenges.
Toxins (Basel) 8:E250.
new inhibitors of SVMPs. Hence, SVMP BaP1 was used as a Bode W, Gomis-Ruth FX, Stockler W. (1993). Astacins, serralysins,
model and two different strategies, a virtual screening on the snake venom and matrix metalloproteinases exhibit identical zinc-
Prestwick Chemical Library, and rational peptide design, binding environments (HEXXHXXGXXH and Met-turn) and
26 L. M. Preciado & J. A. Pereañez Toxin Rev, 2018; 37(1): 19–26

topologies and should be grouped into a common family, the Januário AH, Santos SL, Marcussi S, et al. (2004). Neo-clerodane
‘‘metzincins’’. FEBS Lett 331:134–40. diterpenoid, a new metalloprotease snake venom inhibitor from
Borkow G, Gutiérrez JM, Ovadia M. (1997). Inhibition of the Baccharis trimera (Asteraceae): anti-proteolytic and anti-hemorrhagic
hemorrhagic activity of Bothrops asper venom by a novel neutralizing properties. Chem Biol Interact 150:243–51.
mixture. Toxicon 35:865–77. Katkar GD, Sharma RD, Vishalakshi GJ, et al. (2015). Lupeol derivative
Chinnasamy S, Chinnasamy S, Nagamani S, Muthusamy K. (2014). mitigates Echis carinatus venom-induced tissue destruction by
Identification of potent inhibitors against snake venom metallopro- neutralizing venom toxins and protecting collagen and angiogenic
teinase (SVMP) using molecular docking and molecular dynamics receptors on inflammatory cells. Biochim Biophys Acta 1850:
studies. J Biomol Struct Dyn 33:1516–27. 2393–409.
D’Alessio S, Gallina C, Gavuzzo E, et al. (1999). Inhibition of Lingott T, Schleberger C, Gutiérrez JM, Merfort I. (2009). High-
adamalysin II and MMPs by phosphonate analogues of snake venom resolution crystal structure of the snake venom metalloproteinase
peptides. Bioorg Med Chem 7:389–94. BaP1 complexed with a peptidomimetic: insight into inhibitor
da Silva JO, Fernandes RS, Ticli FK, et al. (2007). Triterpenoid binding. Biochemistry 48:6166–74.
saponins, new metalloprotease snake venom inhibitors isolated from Mahadeswaraswamy YH, Kumar MS, Gowtham YJ, et al. (2011). The
Pentaclethra macroloba. Toxicon 50:283–91. polyphenol 3, 4, 5-tri-hydroxy benzoic acid inhibits Indian Daboia
Dı́az C, Valverde L, Brenes O, et al. (2005). Characterization of events russelli venom and its hemorrhagic complex induced local toxicity.
associated with apoptosis/anoikis induced by snake venom metallo- Curr Top Med Chem 11:2520–30.
proteinase BaP1 on human endothelial cells. J Cell Biochem 94: Markland FS, Swenson S. (2013). Snake venom metalloproteinases.
520–8. Toxicon 62:3–18.
Domingos TFS, Vallim MA, Cavalcanti DN, et al. (2015). Effect of Mazzi MV, Marcussi S, Carlos GB, et al. (2004). A new hemorrhagic
diterpenes isolated of the marine alga Canistrocarpus cervicornis metalloprotease from Bothrops jararacussu snake venom: isolation
against some toxic effects of the venom of the Bothrops jararaca and biochemical characterization. Toxicon 44:215–23.
snake. Molecules 20:3515–26. Mendes MM, Vieira SAPB, Gomes MSR, et al. (2013). Triacontyl
Escalante T, Franceschi A, Rucavado A, Gutiérrez JM. (2000). p-coumarate: an inhibitor of snake venom metalloproteinases.
Effectiveness of batimastat, a synthetic inhibitor of matrix metallo- Phytochemistry 86:72–82.
proteinases, in neutralizing local tissue damage induced by BaP1, a Moura-da-Silva AM, Almeida MT, Portes-Junior JA, et al. (2016).
hemorrhagic metalloproteinase from the venom of the snake Bothrops Processing of snake venom metalloproteinases: generation of toxin
asper. Biochem Pharmacol 60:269–74. diversity and enzyme inactivation. Toxins 8:183.
Escalante T, Rucavado A, Fox JW, Gutiérrez JM. (2011). Key events in Nanjaraj Urs AN, Yariswamy M, Ramakrishnan C, et al. (2015).
microvascular damage induced by snake venom hemorrhagic Inhibitory potential of three zinc chelating agents against the
proteolytic, hemorrhagic, and myotoxic activities of Echis carinatus
metalloproteinases. J Proteomics 74:1781–94.
venom. Toxicon 93:68–78.
Fox JW, Serrano SMT. (2008). Insights into and speculations about
Ozcan T, Akpinar-Bayizit A, Yilmaz-Ersan L, Delikanli B. (2014).
snake venom metalloproteinase (SVMP) synthesis, folding and
Phenolics in human health. Int J Chem Eng Appl 5:393–6.
disulfide bond formation and their contribution to venom complexity.
Patiño AC, Pereañez JA, Núñez V, et al. (2010). Isolation and biological
FEBS J 275:3016–30.
characterization of Batx-I, a weak hemorrhagic and fibrinogenolytic
Fox JW, Serrano SMT. (2010). Snake venom metalloproteinases. In:
PI metalloproteinase from Colombian Bothrops atrox venom. Toxicon
Mackessy SP, ed. Handbook of venoms and toxins of reptiles. Florida:
56:936–43.
CRC Press Boca Raton, 95–113.
Pereañez JA, Patiño AC, Rey-Suarez P, et al. (2013). Glycolic acid
Gâz Florea ŞA, Gâz Florea A, Kelemen H, Muntean D-L. (2016). Snake
inhibits enzymatic, hemorrhagic and edema-inducing activities of
venom metalloproteinases. Acta Med Marisien 62:106–11. BaP1, a P-I metalloproteinase from Bothrops asper snake venom:
Gutiérrez JM, León G, Lomonte B. (2003). Pharmacokinetic-pharma- insights from docking and molecular modeling. Toxicon 71:41–8.
codynamic relationships of immunoglobulin therapy for envenom- Rucavado A, Escalante T, Gutiérrez JM. (2004). Effect of the
ation. Clin Pharmacokinet 42:721–41. metalloproteinase inhibitor batimastat in the systemic toxicity induced
Gutiérrez JM, León G, Rojas G, et al. (1998). Neutralization of local by Bothrops asper snake venom: understanding the role of
tissue damage induced by Bothrops asper (terciopelo) snake venom. metalloproteinases in envenomation. Toxicon 43:417–24.
Toxicon 36:1529–38. Rucavado A, Henrı́quez M, Garcı́a J, Gutiérrez JM. (2008). Assessment
Gutiérrez JM, Lomonte B. (1989). Local tissue damage induced by of metalloproteinase inhibitors clodronate and doxycycline in the
Bothrops snake venoms. A review. Mem Inst Butantan 51:211–23. neutralization of hemorrhage and coagulopathy induced by Bothrops
Gutiérrez JM, Rucavado A, Escalante T, Dı́az C. (2005). Hemorrhage asper snake venom. Toxicon 52:754–9.
induced by snake venom metalloproteinases: biochemical and bio- Soobrattee MA, Neergheen VS, Luximon-Ramma A. (2005). Phenolics
physical mechanisms involved in microvessel damage. Toxicon 45: as potential antioxidant therapeutic agents: mechanism and actions.
997–1011. Mutat Res 579:200–13.
Gutiérrez JM, Rucavado A. (2000). Snake venom metalloproteinases: Srinivasa V, Sundaram MS, Anusha S, et al. (2014). Novel apigenin
their role in the pathogenesis of local tissue damage. Biochimie 82: based small molecule that targets snake venom metalloproteases.
841–50. PLoS One 9:1–11.
Gyawali R, Ibrahim SA. (2014). Natural products as antimicrobial Sunitha K, Hemshekhar M, Gaonkar SL, et al. (2011b). Neutralization of
agents. Food Control 46:412–29. haemorrhagic activity of viper venoms by 1-(3-dimethylaminopro-
Heath EI, Grochow LB. (2000). Clinical potential of matrix metallo- pyl)-1-(4-fluorophenyl)-3-oxo-1,3-dihydroisobenzofuran-5-carbo-
protease inhibitors in cancer therapy. Drugs 59:1043–55. nitrile. Basic Clin Pharmacol Toxicol 109:292–9.
Herrera C, Escalante T, Voisin MB, et al. (2015). Tissue localization and Sunitha K, Hemshekhar M, Santhosh MS, et al. (2011a). Inhibition of
extracellular matrix degradation by PI, PII and PIII snake venom hemorrhagic activity of viper venoms by N-acetyl cysteine: involve-
metalloproteinases: clues on the mechanisms of venom-induced ment of N-acetyl and thiol groups. Curr Top Med Chem 11:2589–600.
hemorrhage. PLoS Negl Trop Dis 9:1–20. Teronen O, Heikkilä P, Konttinen YT, et al. (1999). MMP inhibition and
Hidalgo M, Eckhardt SG. (2001). Development of matrix metallopro- downregulation by bisphosphonates. Ann N Y Acad Sci 878:453–65.
teinase inhibitors in cancer therapy. J Natl Cancer Inst 93:178–93. Vale LH, Mendes MM, Fernandes RS, et al. (2011). Protective effect of
Howes JM, Theakston RDG, Laing GD. (2007). Neutralization of the Schizolobium parahyba flavonoids against snake venoms and isolated
haemorrhagic activities of viperine snake venoms and venom toxins. Curr Top Med Chem 11:2566–77.
metalloproteinases using synthetic peptide inhibitors and chelators. Villalta-Romero F, Gortat A, Herrera AE, et al. (2012). Identification of
Toxicon 49:734–9. new snake venom metalloproteinase inhibitors using compound
Hu J, Van den Steen PE, Sang QX, Opdenakker G. (2007). Matrix screening and rational peptide design. ACS Med Chem Lett 3:540–3.
metalloproteinase inhibitors as therapy for inflammatory and vascular Yip D, Ahmad A, Karapetis CS, et al. (1999). Matrix metalloproteinase
diseases. Nat Rev Drug Discov 6:480–98. inhibitors: applications in oncology. Invest New Drugs 17:387–99.

You might also like