ArticlePDF AvailableLiterature Review

Pathophysiological Effects of Synthetic Derivatives of Polymeric Alkylpyridinium Salts from the Marine Sponge, Reniera sarai

Authors:

Abstract and Figures

Polymeric 3-alkylpyridinium salts (poly-APS) are among the most studied natural bioactive compounds extracted from the marine sponge, Reniera sarai. They exhibit a wide range of biological activities, and the most prominent among them are the anti-acetylcholinesterase and membrane-damaging activity. Due to their membrane activity, sAPS can induce the lysis of various cells and cell lines and inhibit the growth of bacteria and fungi. Because of their bioactivity, poly-APS are possible candidates for use in the fields of medicine, pharmacy and industry. Due to the small amounts of naturally occurring poly-APS, methods for the synthesis of analogues have been developed. They differ in chemical properties, such as the degree of polymerization, the length of the alkyl chains (from three to 12 carbon atoms) and in the counter ions present in their structures. Such structurally defined analogues with different chemical properties and degrees of polymerization possess different levels of biological activity. We review the current knowledge of the biological activity and toxicity of synthetic poly-APS analogues, with particular emphasis on the mechanisms of their physiological and pharmacological effects and, in particular, the mechanisms of toxicity of two analogues, APS12-2 and APS3, in vivo and in vitro.
Content may be subject to copyright.
Mar. Drugs 2014, 12, 2408-2421; doi:10.3390/md12052408
marine drugs
ISSN 1660-3397
www.mdpi.com/journal/marinedrugs
Review
Pathophysiological Effects of Synthetic Derivatives of Polymeric
Alkylpyridinium Salts from the Marine Sponge, Reniera sarai
Marjana Grandič 1 and Robert Frangež 2,*
1 Institute for Hygiene and Pathology of Animal Nutrition, Veterinary Faculty, University of
Ljubljana, Cesta v Mestni log 47, Ljubljana 1000, Slovenia; E-Mail: marjana.grandic@vf.uni-lj.si
2 Institute of Physiology, Pharmacology and Toxicology, Veterinary Faculty, University of Ljubljana,
Gerbičeva 60, Ljubljana 1000, Slovenia
* Author to whom correspondence should be addressed; E-Mail: robert.frangez@vf.uni-lj.si;
Tel.: +386-1-477-91-31.
Received: 17 March 2014; in revised form: 4 April 2014 / Accepted: 4 April 2014 /
Published: 30 April 2014
Abstract: Polymeric 3-alkylpyridinium salts (poly-APS) are among the most studied
natural bioactive compounds extracted from the marine sponge, Reniera sarai. They
exhibit a wide range of biological activities, and the most prominent among them are the
anti-acetylcholinesterase and membrane-damaging activity. Due to their membrane
activity, sAPS can induce the lysis of various cells and cell lines and inhibit the growth of
bacteria and fungi. Because of their bioactivity, poly-APS are possible candidates for use
in the fields of medicine, pharmacy and industry. Due to the small amounts of naturally
occurring poly-APS, methods for the synthesis of analogues have been developed. They
differ in chemical properties, such as the degree of polymerization, the length of the alkyl
chains (from three to 12 carbon atoms) and in the counter ions present in their structures.
Such structurally defined analogues with different chemical properties and degrees of
polymerization possess different levels of biological activity. We review the current
knowledge of the biological activity and toxicity of synthetic poly-APS analogues, with
particular emphasis on the mechanisms of their physiological and pharmacological effects
and, in particular, the mechanisms of toxicity of two analogues, APS12-2 and APS3,
in vivo and in vitro.
Keywords: alkylpyridinium compounds; APS12-2; APS3; cardiotoxicity; hemolysis;
nicotinic acetylcholine receptors; neuromuscular junction; mouse; rat; synthesis
OPEN ACCESS
Mar. Drugs 2014, 12 2409
1. Introduction
Polymeric 3-alkylpyridinium salts (poly-APS) are one of more than 80 biologically active
compounds found in several marine sponges of the order, Haplosclerida [14]. They have been
isolated from crude extracts of the Mediterranean marine sponge, Reniera sarai. Poly-APS have been
reported to comprise two polymers with molecular weights of 5520 and 18,900 Da, corresponding to
29 and 99100 covalently, head-to-tail linked N-butyl-3-butyl pyridinium monomers [5]. In water
solutions, they form larger supramolecular aggregates [57]. However, recent analyses have indicated
that poly-APS are composed of one monomeric species only, with a molecular weight of
5520 Da [8].
Poly-APS are water-soluble compounds with high degrees of association and a broad spectrum of
interesting biological activities [4,6]. These include hemolytic, cytolytic and cytotoxic activities [6],
antifouling [9,10] and antimicrobial properties, including antibacterial [11] and anti-algal activities [12].
Poly-APS are also very potent, irreversible acetylcholinesterase (AChE) inhibitors [1315]. Due to
their ability to induce transient pore formation in biological membranes [16,17], poly-APS have been
used for stable transfection of various mammalian cells with heterologous DNA and, thus, have a
potential in gene therapy [1820]. Moreover, poly-APS exert selective cytotoxicity against non-small
cell lung cancer (NSCLC) cells, which are the most common form of lung cancer, and express
α7-nicotinic receptors [2123]. Cytotoxic concentrations of poly-APS are in the nanomolar range
(0.360.86 nM) [23] and are much lower than the calculated concentrations in blood plasma inducing
toxic and lethal effects after intravenous (i.v.) compound application. Toxic effects on mammals,
arising from poly-APS interference with the cholinergic system, have been observed following
administration of low doses (0.7 mg/kg) of poly-APS. At higher doses, these effects were masked by
the more pronounced lethal activity of the compound related to hemolysis and platelet aggregation.
The half-lethal dose (LD50) of poly-APS in rats has been estimated to be 2.7 mg/kg ([24], reviewed
in [25]). Poly-APS have recently been shown, at a 1 μM concentration, to diminish endothelium-dependent
relaxation of isolated rat thoracic aorta and to significantly decrease coronary flow in the heart [26].
Such biological effects of natural poly-APS and their possible application in the fields of industry
(as components of environmentally friendly antifouling paints) and medicine (as new anti-cholinergic,
transfection and chemotherapeutic agents) have led to the synthesis of several 3-alkylpyridinium
analogues (sAPS) with different degrees of polymerization and different lengths of the constituent
alkyl chains [2729]. The synthesis of structurally well-defined analogues with different chemical
properties and degrees of polymerization has enabled the regulation of the biological activities
of sAPS.
The aim of this review is to summarize current knowledge on the biological activities and toxicity
of sAPS, with particular emphasis on mechanisms of toxicity of two synthetic analogues, APS12-2 and
APS3, in vivo and in vitro.
2. Synthetic Analogues of Polymeric Alkylpyridinium Salts
Their interesting biological effects, their potential use in the pharmaceutical and chemical
industries, coupled with the insufficient quantities of natural poly-APS, have contributed to the
Mar. Drugs 2014, 12 2410
development of new methods for synthesizing poly-APS analogues. This could enable the commercial
production of sAPS with modified characteristics. In 2004, Mancini and colleagues synthesized dimers
and tetramers of 3-alkylpyridinium salts [27]. In 2010, Houssen and colleagues reported a new
protocol enabling synthesis of larger polymers that possess greater biological activities [28]. To
determine how the structure of sAPS influences the biological activities, several sAPS, with various
lengths of the alkyl chain, numbers of pyridinium rings and with different counter ions (bromide or
chloride), have been synthesized.
Figure 1. Synthesis of poly-(1,3-alkylpyridinium) salts. Reagents and conditions: for
R = alkyl chain: (i) HBr, toluene, reflux overnight followed by neutralization to yield
products with X = Br; thionyl chloride, dichloromethane, room temperature to yield
products with X = Cl; (ii) reflux in acetonitrile or methanol (in the presence of a small
amount of KCl for monomeric chloride), followed by microwave irradiation at 130 °C for
the time length stated for each compound under the experimental section. Adapted from
Zovko et al. [29], with permission from © 2012 Elsevier Ltd.
Mar. Drugs 2014, 12 2411
sAPS Synthesis
A method that enables simple, rapid and affordable synthesis of highly purified alkylpyridinium
compounds with a high degree of polymerization was developed [28,29]. Monomers were prepared
according to a small modification of the method described by Davies-Coleman in 1993 [30]. Pyridyl
alcohol was produced by coupling bromo-alcohol with 3-picoline. Bromide monomers were produced
by neutralization of the alcohol treated with hydrogen bromide, while chloride monomers were
produced by reacting the substrate with thionyl chloride. The monomers were further oligomerized in
the presence of acetonitrile and methanol. Polymers were then formed using microwave-assisted
polymerization. Their length depended on the time of irradiation [28,29]. Interestingly, the critical
micelle concentration of selected sAPS (APS7, APS8 and APS12-2) was found to be above 1 mg/mL [31],
e.g., considerably higher than that determined for natural poly-APS [5].
The chemical synthesis of poly-(1,3-alkylpyridinium) salts is shown in Figure 1.
The method is quick, safe, economical, eco-friendly and enables the production of large amounts of
product [32]. Several sAPS have been produced with various degrees of polymerization, different
cations and different lengths of the alkyl chain. Some analogues are mixtures of polymers with
different degrees of polymerization. The basic chemical properties of the most studied sAPS are
presented in Table 1.
Table 1. Basic chemical properties of polymeric 3-alkylpyridinium salts (poly-APS) and
their synthetic analogues.
Compound
No. of Alkyl
C-atoms
No. of Polymers
and Molar Ratio
Molecular Weight
(kDa)
Degree of
Polymerization
Counter Ion
Reference
Poly-APS
8
1
5.52
29
Cl
[6]
APS3
3
2 (9:1)
1.46 (1.2/3.8)
10 and 32
Cl
[29]
APS7
7
2 (2:1)
2.33 (1.4/4.2)
8 and 24
Cl
[29]
APS8
8
1
11.9
63
Br
[28]
APS12
12
1
12.5
51
Br
[28]
APS12-2
12
1
14.7
60
Br
[28]
3. Biological Activities of sAPS
3.1. Hemolytic and Antimicrobial Activity
Like natural poly-APS, the synthetic analogues (sAPS) have structures similar to those of cationic
detergents [33]. The hemolytic activity for both is directly proportional to the length of the alkyl chain
and the degree of polymerization [34,35]. The hemolytic activity of analogues with low molecular
weights is very low or negligible [28,29,31]. The nature of the counter ion does not influence the
hemolytic activity [29]. The electrophysiological effects of mono-, di- and tetra-meric sAPS [27] were
evaluated also on cultured hippocampal neurons [17]. Here, again, low-molecular sAPS were found to
be much weaker pore formers than the natural poly-APS, indicating that the polymerization degree and
the subsequent formation of the supermolecular structure are crucial for the observed membrane activity.
sAPS possess antimicrobial properties and have proven to be more effective against Gram-positive
(S. aureus) than Gram-negative bacteria (E. coli). The latter are more resistant to sAPS action,
Mar. Drugs 2014, 12 2412
probably due to the additional lipopolysaccharide layer on the cells [27,29]. Their antibacterial activity
increases with the increasing number of positive charges and the length of the alkyl chain. sAPS with a
bromide counter ion are more active than sAPS with a chloride counter ion [11,29]. Interestingly, all
sAPS, except APS3, which is the smallest, have higher antibacterial activities than natural poly-APS [29].
Compared with structurally similar compounds, like cetylpyridinium chloride (CPC), which has
minimal inhibitory concentrations (MIC) for S. aureus and E. coli of <1.47 μM and 470 μM, sAPS are
quite effective, their antibacterial activity against E. coli being greater (MICAPS-12-2 = 34.01 μM) and
against S. aureus being comparable (MICAPS-12-2 = 6.8 μM) to that of CPC [29].
sAPS also inhibit the growth of pathogenic fungi; the length of the alkyl chain and the degree of
polymerization are important. APS12-2, the analogue with the longest alkyl chain and the highest
degree of polymerization, has the highest antifungal activity [29]. The effectiveness of several sAPS
has been compared with that of some standard antifungal drugs. The antifungal activity of analogues
APS12-2 and APS3 was similar to that of miconazole, while other antifungal drugs were ten to a
hundred times more effective than sAPS [29]. sAPS, especially those with longer alkyl chains, are also
effective against saprophytic fungi. The oxygen atom in the alkyl chain of APS8 significantly
decreases its effectiveness. However, APS12-3 is appropriate as a biocide for protecting wood against
the fungus, Gloeophyllum trabeum [29]. Finally, sAPS oligomers and polymers have the ability to
effectively inhibit the settling of the marine barnacle, Amphibalanus amphitrite, larvae and are thus
interesting as antifouling agents [36,37].
3.2. Effects of sAPS on Acetylcholinesterase
The most prominent biological activity attributed to sAPS is probably the inhibition of AChE, the
enzyme in the nervous system synapses that hydrolyses the neurotransmitter, acetylcholine (ACh).
Hydrolysis of ACh takes place at the bottom of a 2 nm-deep enzyme active site gorge, where the
anionic site responsible for choline recognition and the catalytic site with its active serine are located.
At the rim of the gorge, there is another binding site for the substrate and other ligands, called the
peripheral anionic site [38]. This is also the binding site for natural poly-APS. The first
non-competitive binding of poly-APS is followed by several successive phases ending in the
irreversible inhibition of the enzyme, which is due to the aggregation and precipitation
of AChE [13,14].
Unlike the natural poly-APS, the time-course of AChE inhibition by sAPS12 and APS12-2 is linear,
showing the reversibility of inhibition [28]. This compounds act as noncompetitive AChE inhibitors,
by binding to the peripheral anionic site and preventing the binding of ACh inside the enzyme gorge. It
is assumed that binding takes place at this site, because the size of the synthetic analogues is too great
to allow entry to the enzyme gorge, as these sAPS are very potent AChE inhibitors, acting in
picomolar concentrations [28]. They could be used in medicine as drugs for treating conditions in
which ACh secretion is reduced, i.e., Alzheimers disease, myasthenia gravis and eye glaucoma [39].
3.3. Antitumor Activity of sAPS
Recent studies with synthetic analogue APS8 have shown that it is a potent inhibitor of α7-nicotinic
receptors, at concentrations of less than 1 nM [40]. Since this concentration is lower than the inhibition
Mar. Drugs 2014, 12 2413
constant for AChE (1.88 nM), APS8 activity is probably due to the inhibition of receptors and not
AChE. APS8 inhibits the growth of various cancer cell lines, like A549 and SKMES-1, but is not toxic
for normal fibroblasts [40]. Moreover, using flow cytometry and differential staining, it was found that
APS8 triggers the apoptosis of cancer cells in a concentration-dependent manner [40]. This effect may
be due to the antagonistic effect of APS8 on α7-nicotinic receptors, which are particularly abundant in
various tumor cells of the respiratory tract in contrast to non-cancer cells [40]. The apoptosis caused by
APS8 involves both intrinsic and extrinsic pathways and is activated by cell stress. In the extrinsic
pathway, the death receptors are involved and are activated after binding certain ligands. A number of
reactions are triggered, ultimately leading to apoptosis [40]. The results suggest that APS8 or similar
compounds could be considered as promising compounds for antitumor drugs development for some
types of lung cancer [40].
The basic biological activities of the most studied sAPS are summarized in Table 2.
Table 2. Biological activities of poly-APS and their synthetic analogues.
AChE InhibitionKi (nM) *
Hemolysis (s1 at 500 nM) **
IC50 for NSCLC (μM) ***
irreversible inhibition
0.05
4.41
85
0
3000
10
0.1
480
1.875
2.6
478
0.036
5.0
470
NSCLC, non-small cell lung cancer; * [28]; ** [31]; *** [41].
4. Toxicity of APS12-2 and APS3
In view of the possible use of sAPS in medicine and the pharmacy setting, it was essential to
evaluate their effects on mammals and to explore the mechanisms of their toxicity. APS12-2 and APS3
are the most studied sAPS. They were chosen for research due to the different mechanisms of their
toxicity and their different chemical properties, which could account for their physiological,
toxicological and pharmacologic activities.
APS12-2 is an analogue with a higher degree of polymerization and a longer alkyl chain, bearing a
bromide counter ion. It is strongly hemolytic and acts as a non-competitive AChE inhibitor. APS3 is
smaller and shorter, with a chloride counter ion. It is non-hemolytic and acts as a competitive AChE
inhibitor. In vivo and in vitro experiments have provided significant data on the possible adverse
effects of APS12-2 and APS3 on the vital functions of mammalian organisms, related to their effects
on organ systems, organs, tissues and cells, as well as on the molecular level, as described below.
4.1. In Vivo Effects of APS12-2 and APS3
Before performing in vivo experiments, the median lethal dose for both sAPS analogues was
estimated in Balb/c mice. Different doses of APS12-2 and APS3 were administered intravenously to
male Wistar rats. Blood pressure, respiratory activity and electrocardiograms (ECG) were monitored.
At the end of each experiment, vital organs were removed for histological analysis. The estimated
median lethal doses for APS12-2 and APS3 in mice were 11.5 and 7.25 mg/kg [8,42]. Compared to
Mar. Drugs 2014, 12 2414
natural poly-APS, with an estimated LD50 in rats of 2.7 mg/kg [24], the toxicity of APS12-2 and APS3
is low. In in vivo experiments, it was found that rats are more sensitive to both analogues than
mice [8,42].
Sublethal effects of APS12-2 in vivo were determined in rats (sublethal doses of four and
5.5 mg/kg) to provide more understanding of the mechanistic specificity of this APS. i.v. application
leading to mild transient bradycardia similar to that described for poly-APS above, but in this case, the
heart rate gradually recovered. Arterial blood pressure (aBP) decreased significantly immediately
following application. This was followed by a transient increase, then finally, a gradual return to the
basal value. The bradycardia produced by the anticholinergic activity of the compounds, the
hyperkalemia or the lung reflexes may be responsible for the reduction in aBP. The subsequent
increase in aBP could be the consequence of a compensatory increased sympathetic tone as a response
to the hypotension or the direct or indirect effect of the substance on peripheral blood vessel resistance.
The fact that no increase in heart beat frequency was observed during the period of transient
hypertension supports this view. Sublethal doses of APS12-2 also caused significant elevation of blood
potassium levels, which could be an important cause of the cardiorespiratory toxicity of APS12-2 [8].
In rats, the death caused by a lethal dose (11.5 mg/kg) of APS12-2 was due to cardiorespiratory
arrest [8]. Since the latter can be produced at plasma potassium concentrations above 10 mM [4345],
the cardiotoxic effects of APS12-2 may be related to its hemolytic activity and hyperkalemia
(10.44 ± 0.44 mM) [8,31]. Respiratory arrest could be produced by the stimulation of juxtapulmonary
capillary receptors in lung parenchyma [46,47]. These receptors are mechano-sensitive and are
therefore activated by conditions, like pulmonary edema, congestion or pulmonary microembolism [48].
This could be the mechanism of respiratory arrest produced by lethal doses of APS12-2. This
explanation is supported by histopathological findings of acute lesions observed in the pulmonary
vessels of rats, the lysis of aggregated erythrocytes within their lumina and pulmonary edema [8].
APS3 was not lethal in experimental rats at doses up to 20 mg/kg and at cumulative doses up to
60 mg/kg. Only transient changes in blood pressure were observed. The serum potassium level was, as
expected, not significantly altered, due to the absence of APS3 hemolytic activity [42]. In vivo
experiments with APS3 further confirmed the putative role of hyperkalemia in the cardiotoxic activity
of APS12-2.
The effects of APS12-2 and APS3 on in vivo measured parameters are summarized in Table 3.
In vivo experiments with APS 12-2 on mice, injected (2.2 μg/kg) intramuscularly at the base of the
tail, showed that it decreased the compound muscle action potential (CMAP) [49]. Similar time- and
dose-dependent reversible effects on CMAP amplitude were observed in mice after administration of
APS3 at sublethal doses (0.33 mg/kg). Administration by i.v. of cumulative doses of APS3 (up to
60 mg/kg) in rats produced dose-dependent inhibition of nerve-evoked muscle contraction with an ID50
of 37.25 mg/kg. Since APS3 is a water-soluble substance composed of two relatively small polymers
in a molar ratio 9:1,with molecular weights (m.w.) of 1.2 and 3.8 kDa, in contrast to APS12-2
(m.w. 17.7 kDa), it can pass the slit-pore in muscle capillary membranes, reach the postsynaptic
membrane of the neuromuscular junction and cause neuromuscular block. The relative permeability of
skeletal muscle capillary pores to substances with molecular weights of 342 and 5000 Da is
0.4 and 0.2.
Mar. Drugs 2014, 12 2415
Table 3. The effects of APS12-2 and APS3 on significant parameters in rats.
Measured Parameters
APS12-2 *
APS3 **
LD50 (mice)
11.5 mg/kg
7.25 mg/kg
ECG (rats)
bradycardia
second degree atrioventricular block
Ventricular extrasystoles
Transient tachycardia
Arterial blood pressure
Steep decrease immediately after application
First a decrease, then an increase above
base-line value
Breathing
Respiratory arrest soon after application
No effect
Biochemical parameters
Statistically significant increase in K+ level
(10.44 ± 0.44 mM)
Statistically significant increase in K+ level
(5.66 ± 0.37 mM)
Muscle contraction
No effect up to 8.6 mg/kg
ID50 = 37.25 mg/kg
LD50, half-lethal dose; ID50, median inhibitory dose; ECG, electrocardiography; * [8]; ** [42].
In contrast to APS3, APS12-2 (at 11.5 mg/kg) produced cardiorespiratory arrest, due to its
hemolytic activity, associated with hyperkalemia [8,42]. A possible in vivo effect of APS12-2 on
skeletal muscle contraction could therefore not be observed in vivo, since APS 12-2 produces cardiac
arrest and the death of experimental animals due to its hemolytic activity and hyperkalemia at much
lower doses (11.5 mg/kg), as expected for a skeletal muscle contraction block (i.e., as shown by the
calculated non-hemolytic median inhibitory dose (ID50) of 37.25 mg/kg for APS3-induced skeletal
muscle contraction in vivo). In addition, the relative permeability of skeletal muscle capillary pores to
substances with an m.w. of approximately 17 kDa (close to that of APS12-2) is ten times lower
(at 0.03) than in APS12-2, so that the diffusion of APS12-2 is expected to be much slower.
4.2. In Vitro Physiological and Pharmacological Effects of APS12-2 and APS3
Based on the structure and anti-AChE activities of APS12-2 and APS3 (both are quaternary
ammonium compounds), effects on neuro-muscular transmission were expected. sAPS are structurally
related to quaternary ammonium compounds, like physostigmine, bis(7)-tacrine and BW284c51, some
of which have dual effects and, in a concentration-dependent manner, inhibit either AChE or nicotinic
acetylcholine receptors (nAChR) [5053]. The effects on neuro-muscular transmission were revealed
by experiments with both analogues on neuromuscular preparation [42,49]. APS12-2 and APS3 block
nerve-evoked isometric muscle contraction in a concentration-dependent manner [42,49]. To determine
their molecular mechanisms of action, the microelectrode technique on mouse hemidiaphragm
preparations was applied in order to study the effects of APS12-2 and APS3 on skeletal muscle fiber
resting membrane potential (RP), miniature endplate potential (MEPP) and evoked endplate
potential (EPP). The direct influence of sAPS analogues on nAChRs expressed on Xenopus oocytes
was also studied. Both analogues decreased the amplitude of EPPs and MEPPs in a
concentration-dependent manner, indicating that their action may be on nAChRs [42,49]. To confirm
the possibility of the direct effects of APS12-2 and APS3 on muscle-type nAChRs at the
neuromuscular junction, experiments were performed on Xenopus laevis oocytes into which Torpedo
(α2β1γδ) muscle-type nAChRs had been incorporated. It was proven that APS12-2 (IC50 = 0.0005 μM)
Mar. Drugs 2014, 12 2416
and APS3 (IC50 = 0.19 μM) effectively block the acetylcholine-evoked current through the
muscle-type nAChRs expressed in oocyte membranes, due to nAChRs inhibition [42,49].
In order to study the effects of APS12-2 and APS3, to better establish the mechanisms of their
cardiovascular effects and to provide more data on mechanism specificity, experiments were
performed on isolated porcine coronary vessels. In contrast to APS3, which displayed no effect,
APS12-2 induced the contraction of coronary ring preparations in a concentration-dependent manner
(at 1.36 to 13.60 μM). Lanthanum chloride, a non-selective cation channel blocker [54,55], and
verapamil, a selective antagonist of L-type voltage-dependent calcium channels [56], completely
abolished the contraction of coronary rings induced by APS12-2. This indicates that, due to increased
Ca2+ influx through the voltage-gated Ca2+ channels, APS12-2 induces vascular smooth muscle
contraction in a concentration-dependent manner. These results show, for the first time, that APS12-2
induces a concentration-dependent contraction of coronary ring preparations. Coronary vasoconstriction, as
well as hyperkalemia, may contribute to the cardiotoxic effects of APS12-2. It is notable that the
maximal final concentration of APS12-2 (13.60 μM) that produces a significant increase in coronary
ring tension in vitro is comparable to the maximal concentration of APS12-2 in blood plasma in vivo
following the administration of one LD50, which produced arrhythmia and cardiorespiratory
arrest [57].
The effects of APS12-2 and APS3 on the in vitro measured parameters are summarized in Table 4.
Table 4. Physiological and pharmacological effects of APS12-2 and APS3 in vitro.
Measured Parameters
APS12-2 *
APS3 **
Effect
IC50
Effect
IC50
Skeletal muscle
contraction
Nerve-evoked
stimulation
Inhibition
0.74 μM
Inhibition
20.3 μM
Direct stimulation
No effect up to
2.72 μM
N/A
No effect up to
20.55 μM
N/A
Pharmacological effect
atropine
No effect up to 80 μM
N/A
No effect up to
80 μM
N/A
neostigmine
No effect up to 1 μM
N/A
No effect up to
1 μM
N/A
3,4-DAP
Stops muscle contraction
blockade
(300 μM)
N/A
Stops muscle contraction
blockade
(300 μM)
N/A
Effect on
RP
No effect up to 3.40 μM
N/A
No effect up to
68.49 μM
N/A
MEPP
Amplitude decrease,
MEPP disappear above
0.68 μM
N/A
Amplitude decrease,
MEPP disappear above
6.85 μM
N/A
EPP
Amplitude decrease
0.36 μM
Amplitude decrease
7.28 μM
nAChRs inhibition
Inhibition
0.0005 μM
Inhibition
0.19 μM
Effect on coronary rings ***
Contraction
(4.113.6 μM)
N/A
No effect up to
137 μM
N/A
N/A, Not applicable; 3,4-DAP, 3,4-diaminopyridine; RP, resting membrane potential; MEPP, miniature endplate potential; EPP,
endplate potential; nAChRs, nicotinic acetylcholine receptors; * [49]; ** [42]; *** [57].
Mar. Drugs 2014, 12 2417
Their hypotensive action, hemolytic activity (of some compounds) and cytotoxic activity may limit
the use of these substances as anti-tumor therapeutics and anti-cholinergic drugs. These effects are
expressed in vitro at very low concentrations of APS12-2. However, relatively high doses of the tested
compounds have to be used to see these effects in vivo, which makes these compounds suitable for
preclinical testing. In conclusion, the in vivo toxicity of APS3 is probably the result of the reversible
antagonistic action of the compound on nAChRs on motor endplates, as shown in in vivo and in vitro
experiments. On the other hand, the toxicity of hemolytically active APS12-2 is probably related to the
high blood potassium levels and cardiac arrest or to its direct functional effects (mechanical
dysfunction) of APS12-2 on the heart conduction system. This remains to be proven. The coronary
vasoconstriction produced by APS12-2 constitutes an important mechanism that can contribute to the
cardiotoxicity of APS12-2. In general, the toxicity of tested sAPS is relatively low, when compared to
that of natural poly-APS. The sAPS, in particular those that are non-hemolytic, are of interest for
preclinical testing as novel lung tumor chemotherapeutics.
5. Conclusion
Synthetic APS exert a wide range of interesting biological activities that can vary according to their
structure. It was shown that some of them inhibit the growth of lung cancer cells lines, either by
inducing apoptosis or by inhibiting cell division. The putative underlying mechanism might be the
block of the cholinergic system, which is physiologically important for lung cancer cells homeostasis.
Therefore, sAPS could be suitable especially as a new class of chemotherapeutic drugs for treating
non-small cell lung cancer. In recent studies, it was shown that sAPS have low toxicity that encourages
their further investigation and testing as anticancer drugs. The antitumor effects of one of sAPS
(APS8) are currently being preclinically evaluated on a lung carcinoma rodent model and show some
very encouraging results. Synthetic APS could also find their use as agents allowing the stable
transfection of cells, which could lead to their potential applications in medicine and cell biology.
Finally, due to their ability to inhibit the settlement of marine organisms to submerged surfaces, they
could be potentially used as active components of antifouling paints.
Acknowledgements
The authors acknowledge the Slovenian Research Agency (research program P4-0053(RF) and
P4-0092 (MG)) for their financial support, Kristina Sepčić for helpful comments on the manuscript and
Roger Pain for the critical reading of the manuscript.
Conflicts of Interest
The authors declare no conflict of interest.
References
1. Almeida, A.M.P.; Berlinck, R.G.S. Alcalóides alquilpiridínicos de esponjas marinhas. Quim.
Nova 1997, 20, 170185.
Mar. Drugs 2014, 12 2418
2. Sepčić, K. Bioactive alkylpyridinium compounds from marine sponges. Toxin Rev. 2000, 19,
139160.
3. Sepčić, K.; Turk, T. 3-alkylpyridinium compounds as potential non-toxic antifouling agents.
In Antifouling Compounds, Progress in Molecular and Subcellular Biology, Subseries Marine
Molecular Biotechnology; Fusetani, N., Clare, A.C., Eds.; Springer-Verlag: Berlin, Germany,
2006; pp. 105124.
4. Turk, T.; Sepčić, K.; Mancini, I.; Guella, G. 3-Aklypyridinium and 3-alkylpyridine compounds
from marine sponges, their synthesis, biological activities, and potential use. In Studies in Natural
Products Chemistry; Rahman, A., Ed.; Elsevier: Amsterdam, The Netherlands, 2008; pp. 355397.
5. Sepčić, K.; Guella, G.; Mancini, I.; Pietra, F.; Dalla Serra, M.; Menestrina, G.; Tubbs, K.;
Maček, P.; Turk, T. Characterization of anticholinesterase-active 3-alkylpyridinium polymers
from the marine sponge Reniera sarai in aqueous solutions. J. Nat. Prod. 1997, 60, 991996.
6. Sepčić, K.; Batista, U.; Vacelet, J.; Maček, P.; Turk, T. Biological activities of aqueous extracts
from marine sponges and cytotoxic effects of 3-alkylpyridinium polymers from Reniera sarai.
Comp. Biochem. Physiol. C Pharmacol. Toxicol. Endocrinol. 1997, 117, 4753.
7. Sepčić, K.; Muševič, I.; Lahajnar, G.; Turk, T.; Maček, P. AFM imaging of surface adsorbed
polymeric 3-alkylpyridinium salts from the marine sponge Reniera sarai. Int. J. Biol. Macromol.
1999, 26, 353356.
8. Grandič, M.; Sepčić, K.; Turk, T.; Juntes, P.; Frangež, R. In vivo toxic and lethal cardiovascular
effects of a synthetic polymeric 1,3-dodecylpyridinium salt in rodents. Toxicol. Appl. Pharmacol.
2011, 255, 8693.
9. Faimali, M.; Sepčić, K.; Turk T.; Geraci S. Non-toxic antifouling activity of polymeric
3-alkylpyridium salts from the Mediterranean sponge Reniera sarai (Pulitzer Finali). Biofouling
2003, 19, 4756.
10. Garaventa, F.; Faimali, M.; Sepčić, K.; Geraci, S. Laboratory analysis of antimicrofouling activity
of Poly-APS extracted from Reniera sarai (Porifera: Demospongiae). Biol. Mar. Mediterr. 2003,
10, 565567.
11. Chelossi, E.; Mancini, I.; Sepčić, K.; Turk, T.; Faimali, M. Comparative antibacterial activity of
polymeric 3-alkylpyridinium salts isolated from the Mediterranean sponge Reniera sarai and their
synthetic analogues. Biomol. Eng. 2006, 23, 317323.
12. Eleršek, T.; Kosi, G.; Turk, T.; Pohleven, F.; Sepčić, K. Influence of polymeric 3-alkylpyridinium
salts from the marine sponge Reniera sarai on the growth of algae and wood decay fungi.
Biofouling 2008, 24, 137143.
13. Sepčić, K.; Marcel, V.; Klaebe, A.; Turk, T.; Šuput, D.; Fournier, D. Inhibition of
acetylcholinesterase by an alkylpyridinium polymer from the marine sponge, Reniera sarai.
Biochim. Biophys. Acta 1998, 1387, 217225.
14. Sepčić, K.; Poklar, N.; Vesnaver, G.; Fournier, D.; Turk, T.; Maček, P. Interaction of
3-alkylpyridinium polymers from the sea sponge Reniera sarai with insect acetylcholinesterase.
J. Protein Chem. 1999, 18, 251257.
Mar. Drugs 2014, 12 2419
15. Garaventa, F.; Piazza, V.; Zovko, A.; Turk, T.; Chelossi, E.; Falugi, C.; Aluigi, M.G.;
Angelini, C.; Trombino, S.; Gallus, L.; et al. Multiple functions of the cholinesterase inhibiting
polyalkylpyridinium salts extracted from the marine sponge, Haliclona sarai. WSEAS Trans. Biol.
Biomed. 2010, 7, 103113.
16. McClelland, D.; Evans, R.M.; Abidin, I.; Sharma, S.; Choudhry, F.Z.; Jaspars, M.; Sepčić, K.;
Scott, R.H. Irreversible and reversible pore formation by polymeric alkylpyridinium salts
(poly-APS) from the sponge Reniera sarai. Br. J. Pharmacol. 2003, 139, 13991408.
17. Koss, D.; Hindley, K.P.; David, K.C.; Mancini, I.; Guella, G.; Sepčić, K.; Turk, T.; Rebolj, K.;
Riedel, G.; Platt, B.; et al. A comparative study of the actions of alkylpyridinium salts from a
marine sponge and related synthetic compounds in rat cultured hippocampal neurons. BMC
Pharmacol. 2007, 7, doi:10.1186/1471-2210-7-1.
18. Tucker, S.J.; McClelland, D.; Jaspars, M.; Sepčić, K.; MacEwan, D.J.; Scott, R.H. The influence
of alkyl pyridinium sponge toxins on membrane properties, cytotoxicity, transfection and protein
expression in mammalian cells. Biochim. Biophys. Acta 2003, 1614, 171181.
19. Scott, R.H.; Tucker, S.J.; Evans, R.M.; MacEwan, D.J.; Jaspars, M.; Sepčić, K. Cell membrane
permeabilisation by alkylpyridinium sponge toxin preparations and their utilisation as novel
transfection reagents. In Proceedings of Neurotox’03, Neurotoxicological Targets from Functional
Genomics & Proteomics, University of Nottingham, Nottingham, UK, 14 September 2003;
Beadle, D.J., Mellor, I.R., Usherwood, P.N.R., Eds.; Society of Chemical Industry (SCI): London,
UK, 2004; pp. 5158.
20. McLaggan, D.; Adjimatera, N.; Sepčić, K.; Jaspars, M.; MacEwan, D.J.; Blagbrough, I.S.;
Scott, R.H. Pore forming polyalkylpyridinium salts from marine sponges versus synthetic
lipofection systems: Distinct tools for intracellular delivery of cDNA and siRNA. BMC
Biotechnol. 2006, 6, doi:10.1186/1472-6750-6-6.
21. Catassi, A.; Servent, D.; Paleari, L.; Cesario, A.; Russo, P. Multiple roles of nicotine on cell
proliferation and inhibition of apoptosis: implication on lung carcinogenesis. Mutat. Res. 2008,
659, 221231.
22. Egleton, R.D.; Brown, K.C.; Dasgupta, P. Nicotinic acetylcholine receptors in cancer: Multiple
roles in proliferation and inhibition of apoptosis. Trends Pharmacol. Sci. 2008, 29, 151158.
23. Paleari, L.; Trombino, S.; Falugi, C.; Gallus, L.; Carlone, S.; Angelini, C.; Sepčić, K.; Turk, T.;
Faimali, M.; Noonan, D.M.; et al. Marine sponge-derived polymeric alkylpyridinium salts as a
novel tumor chemotherapeutic targeting the cholinergic system in lung tumors. Int. J. Oncol.
2006, 29, 13811388.
24. Bunc, M.; Strupi-Šuput, J.; Vodovnik, A.; Šuput, D. Toxic effects of head-to-tail 3-alkylpiridinium
polymers isolated from the marine sponge Raniera sarai in rat. Toxicon 2002, 40, 843849.
25. Turk, T.; Frangež, R.; Sepčić, K. Mechanisms of toxicity of 3-alkylpyridinium polymers from
marine sponge Reniera sarai. Mar. Drugs 2007, 5, 157167.
26. Lunder, M.; Drevenšek, G.; Hawlina, S.; Sepčić, K.; Ziberna, L. Cardiovascular effects induced
by polymeric 3-alkylpyridinium salts from the marine sponge Reniera sarai. Toxicon 2012, 60,
10411048.
Mar. Drugs 2014, 12 2420
27. Mancini, I.; Sicurelli, A.; Guella, G.; Turk, T.; Maček, P.; Sepčić, K. Synthesis and bioactivity of
linear oligomers related to polymeric alkylpyridinium metabolites from the Mediterranean sponge
Reniera sarai. Org. Biomol. Chem. 2004, 2, 13681375.
28. Houssen, W.E.; Lu, Z.; Edrada-Ebel, R.A.; Chatzi, C.; Tucker, S.J.; Sepčić, K.; Turk, T.;
Zovko, A.; Shen, S.; Mancini, I.; et al. Chemical synthesis and biological activities of 3-alkyl
pyridinium polymeric analogues of marine toxins. J. Chem. Biol. 2010, 3, 113125.
29. Zovko, A.; Vaukner Gabrič, M.; Sepčić, K.; Pohleven, F.; Jaklič, D.; Gunde-Cimerman, N.;
Lu, Z.; Edrada-Ebel, R.; Houssen, W.E.; Mancini, I.; et al. Antifungal and antibacterial activity of
3-alkylpyridinium polymeric analogues of marine toxins. Int. Biodeterior. Biodegrad. 2012, 68,
7177.
30. Davies-Coleman, M.T.; Faulkner, D.J.; Dubowchik, G.M.; Roth, G.P.; Polson, C.; Fairchild, C.
A new EGF-active polymeric pyridinium alkaloid from the sponge Callyspongia fibrosa. J. Org.
Chem. 1993, 58, 59255930.
31. Grandič, M.; Zovko, A.; Frangež, R.; Turk, T.; Sepčić, K. Binding and permeabilization of lipid
bilayers by natural and synthetic 3-alkylpyridinium polymers. Bioorg. Med. Chem. 2012, 20,
16591664.
32. Kappe, C.O. Controlled microwave heating in modern organic synthesis. Angew. Chem. Int. Ed.
2004, 43, 62506284.
33. Malovrh, P.; Sepčić, K.; Turk, T.; Maček, P. Characterization of hemolytic activity of
3-alkylpyridinium polymers from the marine sponge Reniera sarai. Comp. Biochem. Physiol. C
Pharmacol. Toxicol. Endocrinol. 1999, 124, 221226.
34. Kondo, T.; Tomizawa, M. Mechanism of hemolysis by cationic surface-active agents. J. Pharm.
Sci. 1969, 58, 13781381.
35. Zaslavsky, B.Y.; Ossipov, N.N.; Rogozhin, S.V. Action of surface-active substances on biological
membranes III. Comparison of hemolytic activity of ionic and nonionic surfactants. Biochim.
Biophys. Acta 1978, 510, 151159.
36. Faimali, M.; Garaventa, F.; Mancini, I.; Sicurelli, A.; Guella, G.; Piazza, V.; Greco, G.
Antisettlement activity of synthetic analogues of polymeric 3-alkylpyridinium salts isolated from
the sponge Reniera sarai. Biofouling 2005, 21, 4957.
37. Piazza, V.; Dragić, I.; Sepčić, K.; Faimali, M.; Garaventa, F.; Turk, T.; Berne, S. Antifouling
activity of synthetic alkylpyridinium polymers using the barnacle model. Mar. Drugs. 2014, 12,
19591976.
38. Quinn, D.M. Acetylcholinesterase: Enzyme structure, reaction dynamics, and virtual
transitionstates. Chem. Rev. 1987, 87, 955979.
39. Cummings, J.L. Cholinesterase inhibitors: A new class of psychotropic compounds. Am. J.
Psychiatry 2000, 157, 415.
40. Zovko, A.; Viktorsson, K.; Lewensohn, R.; Kološa, K.; Filipič, M.; Xing, H.; Kem, W.R.;
Paleari, L.; Turk, T. APS8, a polymeric alkylpyridinium salt blocks α7 nAChR and induces
apoptosis in non-small cell lung carcinoma. Mar. Drugs 2013, 11, 25742594.
41. Zovko, A. Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana,
Slovenia. Personal communication, 2014.
Mar. Drugs 2014, 12 2421
42. Grandič, M.; Aráoz, R.; Molgó, J.; Turk, T.; Sepčić, K.; Benoit, E.; Frangež, R. Toxicity of the
synthetic polymeric 3-alkylpyridinium salt (APS3) is due to specific block of nicotinic
acetylcholine receptors. Toxicology 2013, 303, 2533.
43. Emberson, J.W.; Muir, A.R. Changes in the ultrastructure of rat myocardium induced by
hyperkalaemia. J. Anat. 1969, 104, 411421.
44. Van der Meer, C.; Valkenburg, P.W.; Snijders, P.M. Studies on hyperkalemia as a cause of death
in intestinal ischemia shock in rats. Circ. Shock 1986, 19, 329345.
45. Parham, W.A.; Mehdirad, A.A.; Biermann, K.M.; Fredman, C.S. Hyperkalemia revisited. Tex.
Heart Inst. J. 2006, 33, 4047.
46. McCaffrey, T.V.; Kern, E.B. Laryngeal regulation of airway resistance. II. Pulmonary receptor
reflexes. Ann. Otol. Rhinol. Laryngol. 1980, 89, 462466.
47. Willette, R.N.; Barcas, P.P.; Krieger, A.J.; Sapru, H.N. Pulmonary resistance and compliance
changes evoked by pulmonary opiate receptor stimulation. Eur. J. Pharmacol. 1983, 91, 181188.
48. Paintal, A.S.; Damodaran, V.N.; Guz, A. Mechanism of excitation of type J receptors. Acta
Neurobiol. Exp. 1973, 33, 1519.
49. Grandič, M.; Aráoz, R.; Molgó, J.; Turk, T.; Sepčić, K.; Benoit, E.; Frangež, R. The
non-competitive acetylcholinesterase inhibitor APS12-2 is a potent antagonist of skeletal muscle
nicotinic acetylcholine receptors. Toxicol. Appl. Pharmacol. 2012, 265, 221228.
50. Sherby, S.M.; Eldefrawi, A.T.; Albuquerque, E.X.; Eldefrawi, M.E. Comparison of the actions of
carbamate anticholinesterases on the nicotinic acetylcholine receptor. Mol. Pharmacol. 1985, 27,
343348.
51. Okonjo, K.O.; Kuhlmann, J.; Maelicke, A. A second pathway of activation of the Torpedo
acetylcholine receptor channel. Eur. J. Biochem. 1991, 200, 671677.
52. Ros, E.; Aleu, J.; Gomez de Aranda, I.; Cantí, C.; Pang, Y.P.; Marsal, J.; Solsona, C. Effects of
bis(7)-tacrine on spontaneous synaptic activity and on the nicotinic ACh receptor of Torpedo
electric organ. J. Neurophysiol. 2001, 86, 183189.
53. Olivera-Bravo, S.; Ivorra, I.; Morales, A. The acetylcholinesterase inhibitor BW284c51 is a potent
blocker of Torpedo nicotinic AChRs incorporated into the Xenopus oocyte membrane. Br. J.
Pharmacol. 2005, 144, 8897.
54. Kasai, H.; Neher, E. Dihydropyridine-sensitive and omega-conotoxin-sensitive calcium channels
in a mammalian neuroblastoma-glioma cell line. J. Physiol. 1992, 448, 161188.
55. Hogestatt, E.D.; Andersson, K.E. Mechanisms behind the biphasic contractile response to
potassium depolarization in isolated rat cerebral arteries. J. Pharmacol. Exp. Ther. 1984, 228,
187195.
56. Triggle, D.J. The pharmacology of ion channels: With particular reference to voltage-gated Ca2+
channels. Eur. J. Pharmacol. 1999, 375, 311325.
57. Grandič, M.; Bajuk, B.P.; Sepčić, K.; Košorok, M.D.; Frangež, R. Effects of synthetic analogues of
poly-APS on contractile response of porcine coronary arteries. Toxicol. In Vitro 2012, 27, 627631.
© 2014 by the authors; licensee MDPI, Basel, Switzerland. This article is an open access article
distributed under the terms and conditions of the Creative Commons Attribution license
(http://creativecommons.org/licenses/by/3.0/).
... This means that the activity only increases to a certain tail length and then decreases. The cut-off effect of the membrane active compounds (which penetrate to the membrane and disrupt its architecture) has been well documented [14,19,20,[36][37][38][39][40][41][42][43][44]. ...
... It can only be speculated whether the different courses of dependences of bactericidal activities against S. aureus ATCC 29213/MRSA SA 3202 (MRSA3) from isolates MRSA1/MRSA2 could be due to their differences in the composition of bacterial membranes [55][56][57][58]. The effect of changing the length of the hydrocarbon tail affecting the extent of antimicrobial activity of the compounds has been discussed (e.g., [36,37,[39][40][41][42][43][44]59,60]). The decrease in biological activity at a higher chain length as a cut-off effect is discussed above. ...
... 3202 (MRSA3) from isolates MRSA1/MRSA2 could be due to their differences in the composition of bacterial membranes [55][56][57][58]. The effect of changing the length of the hydrocarbon tail affecting the extent of antimicrobial activity of the compounds has been discussed (e.g., [36,37,[39][40][41][42][43][44]59,60]). The decrease in biological activity at a higher chain length as a cut-off effect is discussed above. ...
Article
Full-text available
Pattern 1-hydroxy-N-(2,4,5-trichlorophenyl)-2-naphthamide and the thirteen original carbamates derived from it were prepared and characterized. All the compounds were tested against Staphylococcus aureus ATCC 29213 as a reference and quality control strain and in addition against three clinical isolates of methicillin-resistant S. aureus (MRSA). Moreover, the compounds were evaluated against Enterococcus faecalis ATCC 29212, and preliminary in vitro cytotoxicity of the compounds was assessed using the human monocytic leukemia cell line (THP-1). The lipophilicity of the prepared compounds was experimentally determined and correlated with biological activity. While pattern anilide had no antibacterial activity, the prepared carbamates demonstrated high antistaphylococcal activity comparable to the used standards (ampicillin and ciprofloxacin), which unfortunately were ineffective against E. feacalis. 2-[(2,4,5-Trichlorophenyl)carba- moyl]naphthalen-1-yl ethylcarbamate (2) and 2-[(2,4,5-trichlorophenyl)carbamoyl]naphthalen-1-yl butylcarbamate (4) expressed the nanomolar minimum inhibitory concentrations (MICs 0.018–0.064 μM) against S. aureus and at least two other MRSA isolates. Microbicidal effects based on the minimum bactericidal concentrations (MBCs) against all the tested staphylococci were found for nine carbamates, while 2-[(2,4,5-trichlorophenyl)carbamoyl]naphthalen-1-yl heptylcarbamate (7) and 2-[(2,4,5-trichlorophenyl)carbamoyl]naphthalen-1-yl (4-phenylbutyl)carbamate (14) demonstrated MBCs in the range of 0.124–0.461 μM. The selectivity index (SI) for most investigated carbamates was >20 and for some derivatives even >100. The performed tests did not show an effect on the damage to the bacterial membrane, while the compounds were able to inhibit the respiratory chain of S. aureus.
... Synthetic analogs of Poly-3-APS, such as APS12-2, APS3 (Grandic et al., 2011(Grandic et al., , 2013, APS8 (unpublished data) and structurally related compounds, are promising new chemotherapeutic agents that exhibit very low in vivo toxicity in experimental animals. Recently, the effects of some of these synthetic analogs possessing potent AChE-inhibitory properties were studied on neuromuscular transmission in skeletal muscle (reviewed in Grandic and Frangez, 2014) to avoid unwanted peripheral side effects that could appear in patients treated with some AChE inhibitors. APS12-2 acts as a potent non-competitive AChE inhibitor with an inhibitory constant (K i ) of 34 × 10 −3 nM (Houssen et al., 2010). ...
... Its application in medicine led to the synthesis of a series of synthetic analogs with different degrees of polymerization and different lengths of alkyl chains (Houssen et al., 2010). Recently, the effects of some of these synthetic analogs due to their AChE inhibitory potency were studied on the skeletal neuromuscular transmission (reviewed in Grandic and Frangez, 2014). Experiments on skeletal neuromuscular preparations revealed that APS12-2, a non-competitive AChE inhibitor which was previously shown to be selectively cytotoxic toward NSLC cells in vitro, is a potent antagonist of skeletal muscle nAChRs (Grandic et al., 2012). ...
Article
The effects of natural polymeric alkylpyridinium salt (nPoly-3-APS), a potent acetylcholinesterase inhibitor isolated from the marine sponge Reniera sarai, were studied on isolated mouse phrenic nerve-hemidiaphragm muscle preparations using electrophysiological approaches. nPoly-3-APS inhibited nerve-evoked isometric muscle twitch and tetanic contraction in a concentration-dependent manner (IC50=29.4μM and 18.5μM, respectively) and produced a 30-44% decrease of directly muscle-elicited twitch and tetanus amplitudes at 54.4μM. Additionally, nPoly-3-APS (9.1-27.2μM) markedly decreased the amplitude of miniature endplate potentials, while their frequency was only affected at the highest concentration used. Endplate potentials were also inhibited by nPoly-3-APS in a concentration-dependent manner (IC50=20.1μM), without significant change in the resting membrane potential of muscle fibers (up to 54.4μM). In conclusion, our results show, for the first time, that nPoly-3-APS preferentially blocks the neuromuscular transmission, in vitro, by a non-depolarizing mechanism. This strongly suggests that the in vivo toxicity of nPoly-3-APS mainly occurs through an antagonist action of the compound on nicotinic acetylcholine receptors of skeletal muscles.
... The divergence in cytotoxicity of APS7-2 and APS8-2 might be attributed to the variations in alkyl chain lengths and the presence of distinct counter ions within their chemical structures (specifically, Br-in APS7-2 and Cl-in APS8-2, respectively) ( Figure 6). This underscores the significance of the chemical structure in determining the cytotoxic potential of these compounds toward cancer cells, aligning with established principles in previous research [25]. ...
... The divergence in cytotoxicity of APS7-2 and APS8-2 might be attributed to the variations in alkyl chain lengths and the presence of distinct counter ions within their chemical structures (specifically, Br-in APS7-2 and Cl-in APS8-2, respectively) ( Figure 6). This underscores the significance of the chemical structure in determining the cytotoxic potential of these compounds toward cancer cells, aligning with established principles in previous research [25]. ...
Article
Full-text available
Nicotine binds to nicotinic acetylcholine receptors (nAChRs) that are overexpressed in different cancer cells, promoting tumor growth and resistance to chemotherapy. In this study, we aimed to investigate the potential of APS7-2 and APS8-2, synthetic analogs of a marine sponge toxin, to inhibit nicotine-mediated effects on A549 human lung cancer cells. Our electrophysiological measurements confirmed that APS7-2 and APS8-2 act as α7 nAChR antagonists. APS8-2 showed no cytotoxicity in A549 cells, while APS7-2 showed concentration-dependent cytotoxicity in A549 cells. The different cytotoxic responses of APS7-2 and APS8-2 emphasize the importance of the chemical structure in determining their cytotoxicity on cancer cells. Nicotine-mediated effects include increased cell viability and proliferation, elevated intracellular calcium levels, and reduced cisplatin-induced cytotoxicity and reactive oxygen species production (ROS) in A549 cells. These effects of nicotine were effectively attenuated by APS8-2, whereas APS7-2 was less effective. Our results suggest that APS8-2 is a promising new therapeutic agent in the chemotherapy of lung cancer.
... However, in addition to therapeutic effects, this group of drugs may also have several adverse effects due to the inhibition of ChE enzymes in non-target tissues, including activation of muscarinic acetylcholine receptors (mAChR) in the central nervous system (tremor, bradycardia), mixed central and peripheral mAChR activation (nausea, vomiting), peripheral stimulation of mAChR (diarrhea), and overstimulation of nicotinic acetylcholine receptors (nAChRs) in the neuromuscular junction (muscle weakness) [34,35]. It is also known that some compounds with an inhibitory effect on ChEs can antagonize nAChRs in the central and peripheral neuromuscular system and cause paralysis of skeletal muscle fibers [36,37]. ...
Article
Full-text available
Enzyme butyrylcholinesterase (BChE) shows increased activity in some brain regions after progression of Alzheimer's disease and is therefore one of the therapeutic targets for symptomatic treatment of this neurodegenerative disorder. The organoruthenium(II) complex [(η 6-p-cymene)Ru(II)(1-hydroxy-3-methoxypyridine-2(1H)-thionato)pta]PF 6 (C1) was designed based on the results of our previous structure-activity studies. Inhibitory activity toward cholinesterase enzymes shows that this complex selectively, competitively, and reversibly inhibits horse serum BChE (hsBChE) with an IC 50 value of 2.88 µM. When tested at supra-pharmacological concentrations (30, 60, 90, and 120 µM), C1 had no significant effect on the maximal amplitude of nerve-evoked and directly elicited single-twitch and tetanic contractions. At the highest tested concentration (120 µM), C1 had no effect on resting membrane potential, but significantly decreased the amplitude of miniature end-plate potentials (MEPP) without reducing their frequency. The same concentration of C1 had no effect on the amplitude of end-plate potentials (EPP), however it shortened the half-decay time of MEPPs and EPPs. The decrease in the amplitude of MEPPs and shortening of the half-decay time of MEPPs and EPPs suggest a possible weak inhibitory effect on muscle-type nicotinic acetylcholine receptors (nAChR). These combined results show that, when applied at supra-pharmacological concentrations up to 120 µM, C1 does not importantly affect the physiology of neuromuscular transmission and skeletal muscle contraction.
... These sessile organisms from the phylum Porifera produce several secondary metabolites with defensive functions against pathogens, predators or for space competition. Many of these bioactive molecules have been used as drug leads 3,4 and some synthetic derivatives have been approved for clinical use or have progressed until Phase II/III clinical trials 5 . ...
Article
The search for compounds capable of targeting early pathological changes of Alzheimer`s disease (AD), such as oxidative stress and neuroinflammation, is an important challenge. Gracilin A derivatives were recently synthesized, using a pharmacophore-directed retrosynthesis strategy, and found to posses potent neuroprotective effects. In this work, the derivatives 21a, 27a, 27b, 29a, 21b, 22 and 23c (1-7) that had demonstrated mitochondrial-mediated, anti-oxidant effects, were chosen. The ability of compounds to modulate the expression of anti-oxidant genes (CAT, GPx, SOD’s and Nrf2) was determined in SH-SY5Y cells, being the simplified derivatives 2 and 3 the most effective compounds. The anti-neuroinflammatory properties of derivatives were assessed in BV2 microglial cells activated with lipopolysaccharide (LPS). Derivatives decreased the release of cytokines (Il-1β, IL-6, GM-CSF and TNF-α) and other damaging molecules (ROS, NO). Compounds also regulated the translocation of Nrf2 and NFκB, and reduced p38 activation. These protective effects were confirmed in a trans-well co-culture with both cell lines, in which derivatives added to BV2 cells increased SH-SY5Y survival. This work provides new results that demonstrate the neuroprotective properties of gracilin A derivatives, making them promising candidate drugs for AD. Particularly, derivatives 2 and 3 showed the greatest potential as lead compounds for further development.
Chapter
The enzyme acetylcholinesterase (AChE) is a serine hydrolase whose primary function is to degrade acetylcholine (ACh) and terminate neurotransmission. Apart from its role in synaptic transmission, AChE has several “non-classical” functions in non-neuronal cells. AChE is involved in cellular growth, apoptosis, drug resistance pathways, response to stress signals and inflammation. The observation that the functional activity of AChE is altered in human tumors (relative to adjacent matched normal tissue) has raised several intriguing questions about its role in the pathophysiology of human cancers. Published reports show that AChE is a vital regulator of oncogenic signaling pathways involving proliferation, differentiation, cell-cell adhesion, migration, invasion and metastasis of primary tumors. The objective of this book chapter is to provide a comprehensive overview of the contributions of the AChE-signaling pathway in the growth of progression of human cancers. The AChE isoforms, AChE-T, AChE-R and AChE-S are robustly expressed in human cancer cell lines as well in human tumors (isolated from patients). Traditionally, AChE-modulators have been used in the clinic for treatment of neurodegenerative disorders. Emerging studies reveal that these drugs could be repurposed for the treatment of human cancers. The discovery of potent, selective AChE ligands will provide new knowledge about AChE-regulatory pathways in human cancers and foster the hope of novel therapies for this disease.
Article
Full-text available
The interest on the Mediterranean sponge Haliclona sarai was raised some years ago by the fact that this sponge appears free from parasites and fouling organisms present in its environment. The study of such a feature was at the beginning due to the interest in finding new and efficient anti-fouling substances for applicative purposes. The characteristic was related to the expression of poly-alkylpyridinium salts (poly-APs), a mixture of two of 3-octylpyridinium polymers, including 29 and 99 monomeric units. The main effect of this compound was represented by the strong specific and non-toxic acetylcholinesterase inhibition in vitro. The substance was first tested for its effect on larval development and settling of incrusting organisms, such as Amphibalanus amphitrite. The experiments confirmed the ability of Poly-APs to prevent settlement of sessile organism, by impinging on the AChE activity. Acetylcholinesterase is an enzyme associated to the cholinergic signal system, but is also involved in cell-to-cell communication driving embryonic development and in the regulation of several cellular features, such as apoptosis and cell movements, and is present in some tumour cells and biopsies. Cytotoxicity tests on immortalized and primary cell lines derived from lung tumour (NSCLC) showed a poly-APS dose-dependent selective reduction of cell viability, statistically significant. The same cells, exposed to the poly-APS salts exhibited a loss in the mitochondrial potential, and positive response to apoptosis assays. What makes the poly-APS salts interesting as anticancer therapy adjuvant is that they, at the concentrations inducing apoptosis in tumour cells, seem to scarcely affect the viability of lymphocytes isolated from healthy patients. In this promising frame, the need emerges for the isolation of synthetic homologs of poly-APS molecules, in order to start a study for the therapeutical application of the drug.
Article
Full-text available
Polymeric alkylpyridinium salts (poly-APS) isolated from the Mediterranean marine sponge, Haliclona (Rhizoniera) sarai, effectively inhibit barnacle larva settlement and natural marine biofilm formation through a non-toxic and reversible mechanism. Potential use of poly-APS-like compounds as antifouling agents led to the chemical synthesis of monomeric and oligomeric 3-alkylpyridinium analogues. However, these are less efficient in settlement assays and have greater toxicity than the natural polymers. Recently, a new chemical synthesis method enabled the production of poly-APS analogues with antibacterial, antifungal and anti-acetylcholinesterase activities. The present study examines the antifouling properties and toxicity of six of these synthetic poly-APS using the barnacle (Amphibalanus amphitrite) as a model (cyprids and II stage nauplii larvae) in settlement, acute and sub-acute toxicity assays. Two compounds, APS8 and APS12-3, show antifouling effects very similar to natural poly-APS, with an anti-settlement effective concentration that inhibits 50% of the cyprid population settlement (EC50) after 24 h of 0.32 mg/L and 0.89 mg/L, respectively. The toxicity of APS8 is negligible, while APS12-3 is three-fold more toxic (24-h LC50: nauplii, 11.60 mg/L; cyprids, 61.13 mg/L) than natural poly-APS. This toxicity of APS12-3 towards nauplii is, however, 60-fold and 1200-fold lower than that of the common co-biocides, Zn- and Cu-pyrithione, respectively. Additionally, exposure to APS12-3 for 24 and 48 h inhibits the naupliar swimming ability with respective IC50 of 4.83 and 1.86 mg/L.
Article
Full-text available
Naturally occurring 3-alkylpyridinium polymers (poly-APS) from the marine sponge Reniera sarai, consisting of monomers containing polar pyridinium and nonpolar alkyl chain moieties, have been demonstrated to exert a wide range of biological activities, including a selective cytotoxicity against non-small cell lung cancer (NSCLC) cells. APS8, an analog of poly-APS with defined alkyl chain length and molecular size, non-competitively inhibits α7 nicotinic acetylcholine receptors (nAChRs) at nanomolar concentrations that are too low to be acetylcholinesterase (AChE) inhibitory or generally cytotoxic. In the present study we show that APS8 inhibits NSCLC tumor cell growth and activates apoptotic pathways. APS8 was not toxic for normal lung fibroblasts. Furthermore, in NSCLC cells, APS8 reduced the adverse anti-apoptotic, proliferative effects of nicotine. Our results suggest that APS8 or similar compounds might be considered as lead compounds to develop antitumor therapeutic agents for at least certain types of lung cancer.
Article
Full-text available
ALKYLPYRIDINE ALKALOIDS FROM MARINE SPONGES. The chemistry of alkylpyridine al- kaloids originating from marine sponges is comprehensively reviewed, with emphasis on their natural occurrence, methods for their isolation, spectroscopic characterization, biological activi- ties e chemical synthesis. A likely chemotaxonomic role is suggested, as markers for sponges of the Order Haplosclerida (Demospongiae).
Article
Nicotinic acetylcholine receptors (nAChRs) constitute a heterogeneous family of ion channels that mediate fast synaptic transmission in neurons. They have also been found on non-neuronal cells such as bronchial epithelium and keratinocytes, underscoring the idea that they have functions well beyond neurotransmission. Components of cigarette smoke, including nicotine and NNK [4-(methyinitrosamino)-l-(3-pyridyl)-l-butanone], are agonists of nAChRs. Given the association of tobacco use with several diseases, the non-neuronal nAChR signaling pathway has considerable implications for cancer and cardiovascular disease. Recent studies have shown that alpha 7 is the main nAChR subunit that mediates the proliferative effects of nicotine in cancer cells. As a result, a7 nAChR might be a valuable molecular target for therapy of cancers such as lung cancer and mesothelioma. Future studies involving the design of nAChR antagonists with improved selectivity might identify novel strategies for the treatment of tobacco-related cancers. Here we review the cellular roles of non-neuronal nAChRs, including regulation of cell proliferation, angiogenesis, apoptosis, migration, invasion and secretion.
Article
The ability of two alkyl pyridinium sponge toxin preparations (poly-APS and halitoxin) to form transient pores/lesions in cell membranes and allow transfection of plasmid cDNA have been investigated using HEK 293 cells. Poly-APS and halitoxin preparations caused a collapse in membrane potential, reductions in input resistance and increased Ca2+ permeability. At least partial recovery was observed after poly-APS application but recovery was more rarely seen with halitoxin. The transfection with plasmid cDNAs for an enhanced green fluorescent protein and human tumour necrosis factor receptor 2 was assessed for both toxin preparations and compared with lipofectamine. Stable transfection was achieved with poly-APS although it was less efficient than lipofectamine. These results show that viable cells transfected with alien cDNA can be obtained using novel transient pore forming alkyl pyridinium sponge toxins and a simple pre-incubation protocol. This provides the first proof principle that pore-forming alkyl pyridinium compounds can be used to deliver cDNA to the intracellular environment without permanently compromising the plasma membrane.
Article
During the last 30 years, a number of simple 3-alkylpyridine and 3-alkylpyridinium (3-AP) compounds have been isolated from marine sponges belonging to the order Haplosclerida, suggesting these compounds as chemical markers for systematic determination of haplosclerid sponges. They were isolated from haplosclerid sponges either as (i) monomers differing in the length, saturation, branching and termination of the alkyl chains, (ii) cyclic or linear oligomers, or (iii) a mixture of high-molecular weight polymers. What the biosynthetic pathways are in marine sponges, and how these compounds can be made by organic synthesis is also an interesting question addressed in our review. In this regard we focus particularly on organic syntheses by which selective polymerization of 3-alkylpyridinium polymers may be achieved. Structural investigation of alkylpyridinium compounds and the role played by mass spectrometry has also been reviewed. In spite of their relatively simple chemical structure, all these compounds exert a broad spectrum of biological activities. More than 40 monomeric 3-AP compounds have been isolated from marine sponges and the majority bear a nitrogenous functionality at the end of the alkyl chain. Almost all reported 3-AP monomers exert moderate cytotoxicity, in the concentration range of a few micrograms per millilitre, against certain transformed cell lines. An example of 3-alkylpyridines is niphatyne A, from the marine sponge of the genus Niphates which exhibits an IC50 of 0.5 μg/mL against several transformed cell lines. The majority of 3-AP dimers and trimers were isolated from haplosclerid sponges in cyclic forms. Haliclamines are a typical representative of this group. They inhibit the division of fertilized sea urchin eggs as well as the growth of several transformed cell lines. On the other hand cyclostellettamines are typical example of cyclic alkylpyridinium dimers that modulate muscarinic receptors. Recently an increasing number of polymeric alkylpyridinium compounds have been isolated from haplosclerid sponges. Halitoxins, amphitoxins and alkylpyridinium salts from Mediterranean sponge Reniera sarai are the most studied. The latter exhibit a plethora of interesting biological activities. For instance, they are able to make pores in membranes through which DNA can be transfected into the cell, show selective cytotoxic activity against certain human cancer cells and possess non-toxic inhibitory antifouling properties against larvae of several important fouling organisms. The biological activities and potential use of alkylpyridinium polymers are discussed.
Article
APS12-2 and APS3 are synthetic analogues of polymeric alkylpyridinium salts (poly-APS) isolated from the marine sponge Reniera sarai. The aim of the present study was to determine the possible direct contractile effects of these two synthetic molecules on coronary arteries, in order partly to explain hemodynamic and cardiotoxic effects of APS12-2 previously observed in in vivo studies and to reveal possible adverse effects on the organism in the case of their clinical use. In contrast to APS3, APS12-2 caused a concentration-dependent vascular smooth muscle contraction of isolated porcine coronary ring preparations in a concentration-range from 1.36 to 13.60 μM. Lanthanum chloride (5 mM) and verapamil (10 μM) completely abolished the APS12-2 evoked contraction of the coronary rings. Pre-incubation with indomethacin (10 μM) had no effect on the contractile responses of coronary ring preparations. These results indicate that APS12-2 contracts vascular smooth muscle in a concentration-dependent manner, due to an increase of Ca(2+) influx through the voltage-gated Ca(2+) channels. Our data show for the first time that APS12-2 induces concentration-dependent contraction of coronary ring preparations, which may contribute to the cardiotoxic effects of APS12-2, in addition to hyperkalemia.