Applied Microbiology and Biotechnology
https://doi.org/10.1007/s00253-018-9467-6
MINI-REVIEW
Cationic surfactants as antifungal agents
M. Elisa Fait 1,2 & Laura Bakas 1 & Graciela L. Garrote 2,3 & Susana R. Morcelle 1,2 & Mario C. N. Saparrat 2,4,5
Received: 24 September 2018 / Accepted: 21 October 2018
# Springer-Verlag GmbH Germany, part of Springer Nature 2018
Abstract
Fungi—in being responsible for causing diseases in animals and humans as well as environmental contaminations in health and
storage facilities—represent a serious concern to health security. Surfactants are a group of chemical compounds used in a broad
spectrum of applications. The recently considered potential employment of cationic surfactants as antifungal or fungistatic agents
has become a prominent issue in the development of antifungal strategies, especially if such surface-active agents can be
synthesized in an eco-friendly manner. In this review, we describe the antifungal effect and the reported mechanisms of action
of several types of cationic surfactants and also include a discussion of the contribution of these surfactants to the inhibition of
yeast-based-biofilm formation. Furthermore, the putative mechanism of arginine-based tensioactive compounds as antifungal
agents and their applications are also analyzed.
Keywords Cationic surfactants . Antifungal activity . Human pathogens . Antifungal mechanism
Introduction
Despite the high incidence of the increasingly diverse array of fungal pathogens in our daily lives and the proportionally enhanced risks of opportunistic fungal infections,
the prevention and treatment options are rather limited
(Hanson 2008). Fungi are mainly associated with
* M. Elisa Fait
fait.mariaelisa@biol.unlp.edu.ar
* Susana R. Morcelle
morcelle@biol.unlp.edu.ar
* Mario C. N. Saparrat
masaparrat@fcnym.unlp.edu.ar
1
Centro de Investigación de Proteínas Vegetales
(CIPROVE-UNLP-Centro Asociado CICPBA), Departamento de
Ciencias Biológicas, Facultad de Ciencias Exactas, Centro Asociado
CIC PBA, UNLP, La Plata, Argentina
2
Consejo Nacional de Investigaciones Científicas y Técnicas
(CONICET), Buenos Aires, Argentina
3
Centro de Investigación y Desarrollo en Criotecnología de Alimentos
(CIDCA-CONICET-UNLP), La Plata, Argentina
4
Instituto de Fisiología Vegetal (INFIVE-CONICET-UNLP) and
Instituto de Botánica Carlos Spegazzini, Facultad de Ciencias
Naturales y Museo, UNLP, La Plata, Argentina
5
Cátedra de Microbiología Agrícola, Facultad de Ciencias Agrarias y
Forestales, UNLP, La Plata, Argentina
surfaces’ contamination and spoilage of pharmaceutical,
cosmetic, and food products (Sandle et al. 2014).
Contaminated environmental surfaces provide an important potential source for transmission of not only many
health care-associated fungal pathogens, but also those
found in recreational public facilities, such as swimming
pools and showers. Spores—being able to persist on the
environment for long periods of time—can be identified
as the main structures responsible for the conquest of new
habitats and substrates (Mallo et al. 2017). In this context,
environmental disinfection of surfaces, equipment, and
devices can be identified as a crucial intervention in the
prevention and control of transmission of potentially infectious microorganisms.
Environmental cleaning can reduce contamination on surfaces. However, less than 50% of hospital room surfaces are
adequately cleaned and disinfected when chemical germicides
are used (Weber and Rutala 2013). Biocides used in pharmaceutical industries and health facilities for the disinfection of
medical devices and surfaces of cleanrooms must have a wide
spectrum of activity. They must also effectively kill the common types of cleanroom environmental isolates and pathogens, including Staphylococcus, Micrococcus, Bacillus,
Penicillium, Cladosporium, and Aspergillus (Sandle et al.
2014). However, among the different microorganisms isolated
from cleanrooms, fungi have received less attention than bacteria and the eradication of fungal contaminants in our immediate environment has been found to be an arduous task, and
Appl Microbiol Biotechnol
many times these practices have been rather a response to an
outbreak than a stand-alone activity (Dancer 2009).
Among the ca. five million species of fungi estimated to
currently exist, approximately 300 have been recorded to
cause disease in humans, but only 20–25 of these do so frequently (Perfect 2017). Within these species, those responsible for superficial fungal infections—in which the pathogen is
restricted to the stratum corneum, with little or no tissue reaction—belong to five major fungal phyla (Table 1). Crosstransmission occurrences of these pathogens are mainly indirect through desquamated epidermis or hairs and environmental surfaces, or direct through bodily contact (Hay 2017). For
treatment, topical agents—formulated as creams, lotions, or
gels—are conventionally preferred, since the side effects are
fewer than their internally taken counterparts (Bseiso et al.
2015). However, growing strain resistance has decreased the
efficacy of many known and commonly used antifungals,
such as fluconazole (Obłąk et al. 2013). To overcome this
situation, a new trend in management of fungal infections
has considered the use of surface-active agents depending
upon their safety—such as the commercial detergent
cetrimide, which has proven to be efficient against fungal
keratitis (Mahmoud 2016).
In this review, we discuss the use of cationic surfactants as
agents for the prevention of fungal colonization and for the
control of fungal environmental contaminations in surfaces
and devices, providing also an insight into the main mechanisms involved in these features. Furthermore, we will illustrate the role of model-membrane systems in the study of the
interaction of bioactive compounds with the fungal membrane
as the main target. These kinds of data can become a powerful
tool for medicinal chemistry and pharmaceutical technology
through the design and optimization of the antifungal activity
of novel compounds that exert their activity at surface level.
What do we know about surfactants?
The term surfactant has its origins in the combination of the
words surface-active agent, whereas the suffix -ant refers to
performing a specific action. According to the Encyclopædia
Britannica, surfactants are compounds that, when added to a
liquid, reduce its surface tension. From the historical point of
view, surfactant compounds were originally intended for
cleaning. At the present time, however, these agents possess
a plethora of applications, from being general detergents (as
with sodium lauryl ether sulfate) up to acting as specific biocides that are never used as detergents (such as the fungicide
dodine).
In general, surfactants are ingredients in many products
used in daily life—such as cleaners (soaps and detergents for
industrial, institutional, and home use), pharmaceutical formulations, food, agrochemicals, plastics, personal care, and
cosmetics, among others. Because of their ability to reduce
the surface tension between immiscible systems (liquid/liquid
or solid/liquid), surfactants are mainly used as emulsifiers,
dispersants, solubilizers, and wetting and foaming agents.
These properties are based on their amphiphilic nature: surfactant molecules have two main functional moieties, one polar (i.e., water-miscible), the other nonpolar (i.e., oil-miscible).
Owing to their numerous applications, surfactants are
chemicals that experience a great demand worldwide. The
global requirement for these compounds was 15.9 million tons
in 2014 and is expected to reach 24 million tons by 2022
(Grand View Research 2016). The world market for surfactants was estimated at 30.7 billion dollars in 2015 and at an
annual growth of around 5%. Consequently, the market forecast is expected to reach 45.0 billion dollars by 2024 (Acmite
Mark 2016). This market boosting is mainly a result of the
rising concerns of people regarding health and personal care
all over the world (6.3% from 2015 to 2022); other influences
include the increasing demand for oil-field chemicals, as well
as other technological innovations (Occams Business
Research & Consulting 2017).
According to the combined hydrophilic and hydrophobic
handles of the molecule, surfactants can be classified as anionic, cationic, zwitterionic (amphoteric), or nonionic
(Florence and Atwood 2006). Among all these classes of surfactant, the anionic ones are by far the more widely used, thus
holding the largest market share, mainly for their use as household cleaners and certain pharmaceutical formulations. In general, the anionic surfactants are sulfonates: linear
alkylbenzene, secondary alkane, alpha-olefin, and methyl ester sulfonate, with the first of those being the most commonly
used for detergents and other cleaners for the last 30 years
(Hayes 2009; Gong et al. 2016). Zwitterionic, or amphoteric,
surfactants may develop a positive or negative net charge depending on the pH. This class has low foaming characteristics
and good wetting properties, imparting mildness to personal
care formulations especially. The alkyl betaines as well as the
alkyl amidopropyl betaines are the most representative among
this type of compound (Johansson and Somasundarau 2007).
Nonionic surfactants are mainly represented by alcohol
ethoxylates; this class is used principally for defoaming and
as solubilizing agents in pharmacons (e.g., polysorbates and
sorbitan esters among others; Florence and Atwood 2006;
Johansson and Somasundarau 2007). Finally, among the four
classes of surfactants, the cationic group is expected to be the
faster to grow in the world market in the near future, primarily
owing to the multifunctional role of these surfactants in cosmetics and pharmaceutical formulations (Occams Business
Research & Consulting 2017).
Among the different parameters that can be taken into consideration to classify these compounds, surfactants can be also
categorized according to their origin into synthetic surfactants
or natural class—also known as biosurfactants—, the latter
Examples of fungal species responsible of human superficial infections
Phylum*
Subphylum*
Class*
Order*
Genus*
Basidiobolomycota
Basidiobolomycotina
Basidiobolomycetes
Basidiobolales
Basidiobolus
Entomophthoromycota Entomophthoromycotina Entomophthoromycetes Entomophthorales Conidiobolus
Mucoromycota
Mucoromycotina
Mucoromycetes
Mucorales
Apophysomyces,
Mucor, and
Rhizopus
Ascomycota
Saccharomycotina
Saccharomycetes
Saccharomycetales Candida
Pezizomycotina
Dothideomycetes
Pleosporales
Alternaria
Eurotiomycetes
Capnodiales
Chaetothyriales
Hortaea
Cladophialophora
Exophiala
Eurotiales
Onygenales
Basidiomycota
Fonsecaea
Phialophora
Rhinocladiella
Aspergillus
Penicillium
Coccidioides
Epidermophyton
Hystoplasma
Microsporum
Paracoccidioides
Trichophyton
Sordariomycetes
Hypocreales
Fusarium
Ustilaginomycotina
Malasseziomycetes
Microascales
Ophiostomatales
Sordariales
Malasseziales
Pseudallescheria
Sporothrix
Madurella
Malassezia
Agaricomycotina
Tremellomycetes
Trichosporonales
Trichosporon
*Taxonomic levels are defined according to Tedersoo et al. (2018) and MycoBank (2018)
Species*
Type of mycosis
B. ranarum
C. coronatus, C. incongruus
R. oryzae
Subcutaneous and gastrointestinal disease
Central facial disease
Cutaneous mucormycosis
C. albicans, C. glabrata, C. parapsilosis,
and C. tropicalis
A. alternate
Superficial candidiasis
Keratitis, ulcers, cysts, opportunistic human
infections, allergic bronchopulmonary mycosis
H. werneckii
Tinea nigra
C. bantiana
Brain abscesses, keratitis, ulcers, cysts, cerebral
phaeohyphomycosis
E. dermatitidis
Subcutaneous phaeohyphomycoses after traumatic
implantation, keratitis, pneumonia, otitis
F. pedrosoi
Chromoblastomycosis
P. verrucosa
Chromoblastomycosis
R. similis
Mycetoma and chromoblastomycosis
A. fumigatus and other Aspergillus species Superficial aspergillosis (mainly A. fumigatus),
otomycosis, keratitis, and onychomycosis
P. marneffei
Keratitis and otomycosis
C. immitis
Valley fever
E. floccosum
Tinea
H. capsulatum
Histoplasmosis
M. canis, M. audouinii, and M. gypseum
Tinea
P. brasiliensis
Paracoccidioidomycosis
T. rubrum, T. tonsurans, T. mentagrophytes, Tinea
T. verrucosum, and T. schoenleinii
F. solani
Opportunistic skin infections, mycetoma, human
fusariosis, keratitis
P. boydii
Mycetoma
S. schenckii
Sporotrichosis
M. mycetomatis
Black-grain mycetoma
M. furfur, M. globosa, and M. sympodialis Pityriasis versicolor, seborrhoeic dermatitis, and
folliculitis
T. asahii, T. ovoides, T. inkin, and
Trichosporosis (white and black piedra),
T. mucoides
opportunistic pathogen
Appl Microbiol Biotechnol
Table 1
Appl Microbiol Biotechnol
being produced by living organisms, mainly microorganisms
(Tripathy et al. 2018). Biosurfactants consist of glycolipids,
neutral lipids, and lipopeptides, as well as other molecules of
larger molecular mass such as lipoproteins and complexes
formed by lipopolysaccharides and proteins in addition to
complexes of polysaccharides, lipids, and proteins (Hayes
2009). Synthetic equivalents to biosurfactants can be designed
imitating natural amphiphilic structures such as phospholipids, alkyl glucosides, and acyl amino acids. This review
will focus on synthetic cationic surfactants, particularly on
those based on amino acids, and their use as antifungal agents.
Can cationic surfactants help to control fungal
environmental and superficial
contaminations?
We need to remark here that the amino acid-based surfactants
are not commonly used as detergents, since these compounds
exhibit a lower foaming capacity than their anionic counterparts (Johansson and Somasundarau 2007). Nevertheless, this
class has many other applications—e.g., a retardation of steel
corrosion in strongly acidic media (Aiad et al. 2014a; Shaban
et al. 2015a, b; Hegazy et al. 2016) and the removal of heavy
metals (chromium and arsenic) that are present in industrial
wastes like oxyanion environmental contaminants (Li et al.
2002; Li et al. 2003; Gecol et al. 2004) along with environmental uses such as functioning as flushing additives for clearing Cs+ and radionuclides from contaminated soils (Mao et al.
2015), assisting in the dewatering of activated sludge (Wang
et al. 2014), and enhancing oil recovery (through a tailor-made
mixture of cationic and anionic surfactants) by reducing crude
oil-water interfacial tension and thus producing a flow of oil,
among others (Li et al. 2014; He and Xu 2017).
Notwithstanding, apart from this compatibility with industrial and environmental applications, the most relevant property of cationic surfactants with respect to the enhancement of
human well-being is their biocidal ability against a wide range
of microorganisms. This feature together with their amphiphilic nature makes them powerful additives as well as active
compounds by themselves for use in different pharmaceutical,
medical, and cosmetic products, though up to now cationic
surfactants have been used traditionally as disinfectants
(Florence and Atwood 2006; Johansson and Somasundarau
2007). The most well-known compounds of this kind are the
quaternary ammonium salts (QUATs, also known as QACs)
and esterquats. QACs were introduced in the late 1930s and
are considered as high-production-volume chemicals. For example, these kinds of compounds are present in about 36% of
disinfectant formulations. Their chemical structure is
R1R2N’R3R4, (where the Rs are alkyl groups), and they are
used mainly in disinfectants and antiseptic formulations of
household, industrial, and institutional cleaners; in human
and animal health care preparations; and as in agricultural
and industrial facility products. Besides their broad antimicrobial spectrum at low concentrations, QACs have many other
advantages, such as no color, low odor, high stability, compatibility with the other ingredients in several formulations, and
relatively low toxicity (Tezel and Pavlostathis 2015). Within
this context, QACs are biodegradable under aerobic conditions and consequently are present in surface waters and sediments in concentrations below their minimum inhibitory concentration (MIC), a property that could lead to the emergence
of resistant bacterial strains, including those of certain pathogenic genera, such as the already documented case of
Staphylococcus aureus (Tezel and Pavlostathis 2015;
Jennings et al. 2015). QACs are also toxic to aquatic organisms including algae, fish, crustaceans, and protozoans plus
other microorganisms (Chen et al. 2014; Lavorgna et al. 2016;
Di Nica et al. 2017). Moreover, though the QACs are extensively used in personal care formulations, those surfactants
can prove to be irritants, occasioning different types of dermatitis as well as other allergic eruptions (Anderson et al. 2016;
Isaac and Scheinman 2017). For all these reasons, the design
and production of alternative cationic surfactants is a topic of
utmost interest.
The search for novel cationic
surfactants—from those with eco-friendly
characteristics to those with improved
physicochemical and biologic properties
Bio-based surfactants are all those composed either partially
or totally of biologic products (namely, renewable material of
agricultural and/or forestry origin) whose production, usage,
and disposal have low impact to both the users and the environment (Hayes 2009). Both the safety and the environmental
and health profiles of these kinds of products make them not
only more attractive to consumers who are concerned about
the urgency of ecological issues, but also imperative to any
potential users who need to meet the more restrictive requirements and standards exacted by most of the regulations imposed by many governments worldwide (Jessop et al. 2015).
The biosurfactants mentioned earlier fall into this category of
compounds.
Certain biosurfactants can be taken as models for the synthesis of novel structures. This application has been especially
explored in the example of lipoaminoacids and their analogues, all of which compounds can be found in cell membranes. Owing to their structural simplicity, these molecules
are relatively easy to design and to synthesize, even in terms of
green chemistry criteria. Synthons for the production of these
kinds of compounds are proteinogenic and nonproteinogenic
amino acids for the polar moiety of the molecule and fatty
amines or fatty alcohols for the hydrophobic residues
Appl Microbiol Biotechnol
(Infante et al. 2009). Among the plethora of amino acid-based
surfactants that can be found in the literature (Gerova et al.
2008; Pérez et al. 2009; Pang and Chu 2010; Tripathy et al.
2018), the arginine-based surfactants are among those with the
most striking properties, mainly given by the guanidinium
group present in the side chain of the amino acid, which moiety is positively charged at neutrality but also even at high pH
levels. Comprehensive reviews can be found on the physicochemical and biologic features of amino acid- and argininebased surfactants in particular (Pinazo et al. 2011; Lozano
et al. 2011; Chandra and Tyagi 2013; Singh and Tyagi 2014;
Bordes and Holmberg 2015; Pinazo et al. 2016; Tripathy et al.
2018). The arginine-based ones are especially promising in
view of their antimicrobial properties, extremely low toxicity,
low irritation potential, and more facile biodegradability than
that of the QACs. In this regard, the ethyl ester of N-lauroyl-Larginine, commercially known as Mirenat®, is an argininebased surfactant commonly used as an emulsifier and a preservative in different food products (Terjung et al. 2014; Maier
et al. 2014; Manrique et al. 2017; Gaikwad et al. 2017).
Despite the actual uses as well as other potential applications
that these kinds of compounds may have, such as nanocarriers
for gene therapy (Rosa et al. 2007; Jiang et al. 2016; Peña et al.
2017) and drug delivery systems (Tavano et al. 2014;
Nogueira et al. 2015), their biocidal activity is as yet incompletely studied since their antifungal action was only investigated against certain Candida species and other specific
human-pathogenic fungal strains (Morán et al. 2001; Castillo
2006; Colomer et al. 2011; Fait et al. 2018).
Antifungal activity of cationic surfactants
Table 2 summarizes the biocidal effects of cationic surfactants
on several fungi and oomycota that have been reported up to
the present. Most findings on the antifungal activity of cationic surfactants are with respect to the fungi that belong to the
Ascomycota phylum as well as certain species from the phyla
Basidiomycota (e.g., dodemorph acetate, guazatine, lauric
arginate) and Mucoromycota (e.g., guazatine, lauric arginate)
along with those from the phylum Oomycota (Kingdom
Straminipila; e.g., dodine).
Within the context of biologic infections, the central relevance of biofilms comes from their relationship to infections
associated with devices of hospital use, such as intravenous
and urethral catheters; permanent prostheses, dental or other
types; and mechanical heart valves (Desai et al. 2014). Not
only can these infections arise from the microbial colonization
of the surfaces of the devices and their growth in biofilms, but
the detachment of cells from those biofilms can also cause
even more severe infections and septicaemia (Jabra-Rizk
et al. 2004). So far, only echinocandins and liposomal formulations of amphotericin B have displayed a significant activity
against fungal biofilms (Tsui et al. 2016). In fact, the adherence of pathogenic microorganisms to surfaces and tissues—
the first step in the formation of biofilms—happens to be an
excellent target for antifungal therapies, and a study of the
antiadhesive and inhibitory properties of biofilm formation
exhibited by compounds with antifungal activity is of utmost
relevance.
Dusane et al. (2012) demonstrated that rhamnolipids are
efficient biologic surfactants for disrupting preformed
biofilms by the yeast Yarrowia lipolytica, reducing those
structures by 46% at concentrations below the MIC, with sodium dodecyl sulfate and cetrimonium bromide (CTAB) being less effective at 38 and 25%, respectively. Regarding cell
adhesion, although CTAB was reported to bind to the negatively charged microbial surfaces, alter the surface charge, and
prevent the binding of fungal cells to those surfaces, this surfactant was not as effective as rhamnolipids in preventing
Y. lipolytica adhesion, with the antiadhesive effect of
rhamnolipids being significantly higher at an inhibition of
adhesion to microtiter plate wells by 50% at the MIC.
Candida albicans—the predominant causal agent of human
candidiasis—possesses various virulence attributes including
the property of biofilm formation. Candida biofilms have
been reported to be 30–2000 times more resistant to various
antifungal agents than their planktonic counterparts (Hawser
and Douglas 1994). Given the increased resistance of such
pathogenic microorganisms to the currently used antibiotics
and chemotherapeutic agents, natural products such as 4hydroxycordoin—it was derived from plants—constitute an
alternative for the prevention and treatment of such infections
(Messier et al. 2011). Farnesol also has been described as
acting as a naturally occurring quorum-sensing molecule that
inhibits biofilm formation, thus indicating the potential of this
natural intermediate in the biosynthesis of cholesterol for further development and use as a novel therapeutic agent
(Ramage et al. 2002).
Holtappels et al. (2017) investigated the influence of
oleylphosphocholine (OlPC) on three different developmental
stages of biofilms on catheters—inhibition of cellular adhesion and/or biofilm development, and disruption of preformed
biofilms—of 14 strains and clinical isolates of C. albicans.
This study demonstrated that, although OlPC had no effect
on C. albicans adhesion, the biofilm development was significantly reduced at low concentrations, thus evidencing also
changes in biofilm architecture, as confirmed by scanning
electron microscopy. The thick layer of hyphal cells embedded in the material present in nontreated wild-type cells was
replaced by a rudimentary biofilm composed of hyphal cells
attached to a substrate, and, in strikingly impressive effectiveness, higher concentrations of OlPC completely abolished
biofilm development. The authors also investigated the activity of OlPC on in vivo biofilms of C. albicans developed in a
rat subcutaneous biofilm model, demonstrating that daily oral
Appl Microbiol Biotechnol
Table 2
Reported effects of cationic surfactants on fungi and oomycota
Surfactant*
Fungal taxa and Oomycota tested/susceptibility**
Reference
Cetyltrimethylammonium bromide [CTAB]
Complete inhibition of A. ochraceusa conidia after 3 days
when exposed at 0.5%. C. kruseia FR 01190 was killed
within 15 min of contact when applied to 0.5%. While a
few cells of C. parapsilosisa FR-01760 survived if the
contact time was less than 60 min, C. albicansa FR-00806
had no effect when applied at 0.5%.
A. flavusa (MIC of0.01 mg/mL), F. solania (MIC of
100 μg/mL).
A. flavusa (MIC90 of 8 μg/mL), A. fumigatusa (MIC90 of
16 μg/mL), A. nigera (MIC90 of 8 μg/mL), Penicilliuma
spp. (MIC90 of 16 μg/mL), Fusariuma spp. (MIC90 of
8 μg/mL), Cladosporiuma spp. (MIC90 of 8 μg/mL),
Curvulariaa spp. (MIC90 of 8 μg/mL), and Alternariaa
spp. (MIC90 of 16 μg/mL)
Baker’s yeast, S. cerevisiaea
Several fungi such as A. nigera, A. ochraceusa, F. solania,
P. minioluteuma, and T. harzianuma were not able to
sporulate in vitro at 0.06%. Except for A. nigera, their
growth rate was also reduced, but all these fungi were
completely inhibited at 0.07%. The B. bassianaa growth
rate was strongly inhibited when applied to 0.06%.
50% viability of C. albicansa ATCC 90028 at 0.3 mM.
Gupta et al. 2002
Dodemorph acetate [Meltatox]
Dodecylguanidinium acetate [dodine]
Nα, Nx-bis (Nα-lauroylarginine)-a,x-alkylidenediamide
[bis(Args)]
Sugar-based gemini cationic amphiphiles
C. gloeosporioidesa (IC50 of the growth: 73.2 μM),
C. lindemuthianuma (IC50 of the growth: 79.1 μM),
F. oxysporuma (IC50 of the growth: 80.8 μM), F. solania
(IC50 of the growth: 67.9 μM), T. rubruma (IC50 of the
growth: 51.48 μM; MIC7: 62.5 μM; MIC21: 125 μM),
and T. mentagrophytesa (IC50 of the growth: 37.19 μM;
MIC7: 62.5 μM; MIC21: 62.5 μM).
S. pannosaa var. rosae (recommended dosage for its control,
200 μg/mL), Erysiphea species, and S. flocculosaa ATCC
74320
Although the B. bassianaa growth rate was strongly inhibited
when applied at 0.046% (95%), the surfactant inhibits
sporulation of M. anisopliaea.
When applied as the commercial funguicide, Efuzin 500FW
at practical field dose (0.8–1 L/ha) caused complete
inhibition of germination of conidia of C. acutatuma, a
causal agent of anthracnose of sour cherry.
S. graminicolad, pearl milletdowny mildew
The surfactant selectively blocked the growth of A. nigera,
A. flavusa, C. cladosporioidesa, C. clavisporusa,
F. roseuma, H. thompsoniia, N. rileyia, and Sporothrix
insectoruma when applied at 50 mg/L.
A. nigera (MIC, 0.10 mM for a gemini compound with two
symmetrical C16-long chain groups linked by a spacer
chain of C2 or C4; 0.19 mM for a gemini compound with
two symmetrical C16-long chain groups linked by a spacer
chain of C6; 0.24 mM for a gemini compound with two
symmetrical C12-long chain groups linked by a spacer
chain of C2; 0.16 mM for a gemini compound with two
symmetrical C12-long chain groups linked by a spacer
chain of C4) and C. albicansa.
A. nigera (24.5 mm of IZD by a dodecyl derivative of
glucose-based surfactant at 5 mg/mL; 11 mm of IZD by an
oleate derivative of glucose-based surfactant at 5 mg/mL;
12 mm of IZD by a dodecyl derivative of fructose-based
surfactant at 5 mg/mL; 9 mm of IZD by an oleate
Mahmoud 2016
Sandle et al. 2014
Raicu 1998
Posadas et al. 2012
Vieira and
Carmona-Ribeiro
2006
Fait et al. 2018
Benyagoub and
Bélanger 1995
Posadas et al. 2012
Tóth et al. 2012
Deepak et al. 2006
Luz et al. 2007
Pérez et al. 1996; Tyagi
and Tyagi 2014
Negm and Mohamed
2008
Appl Microbiol Biotechnol
Table 2 (continued)
Surfactant*
Octamethylenediamine,
iminodi(octamethylene)diamine,
octamethylenebis(imino-octamethylene)diamine,
and carbamonitrile [guazatine]
Nα-benzoyl-arginine decylamide [Bz-Arg-NHC10]
Nα-benzoyl-arginine dodecylamide [Bz-Arg-NHC12]
Lauramide of L-arginine ethyl ester
monohydrochloride; ethyl-N″-lauroyl-L-arginate
HCl [Lauric arginate, LAE, Mirenat®]
Nα-Lauroyl arginine methylester (LAM)
Nα-Nonanoyl L-Arginine ethyl ester [NAE]
Nα-Myristoyl-L-Arginine ethyl ester [MAE]
Myristamidopropyl dimethylamine [MAPD]
Fungal taxa and Oomycota tested/susceptibility**
derivative of fructose-based surfactant at 5 mg/mL) and
A. flavusa (18 mm of IZD by a dodecyl derivative of
glucose-based surfactant at 5 mg/mL; 25 mm of IZD by an
octadecyl derivative of glucose-based surfactant at
5 mg/mL; 9 mm of IZD by an oleate derivative of
glucose-based surfactant at 5 mg/mL; 12.7 mm of IZD by
a dodecyl derivative of fructose-based surfactant at
5 mg/mL; 12 mm of IZD by an oleate derivative of
fructose-based surfactant at 5 mg/mL)
Several isolates of P. digitatuma (3.1 ppm producing an
inhibition growth rate of 1.8% to 4.9%), G. citri-aurantiia
(isolate INTA 8 growth inhibited by 4.9% by 3.1 ppm and
by 12.6% by 75 ppm).
Several Candidaa strains, including fluconazole-resistant
clinical isolates of C. albicansa, C. kruseia, C. glabrataa,
and C. tropicalisa (MIC50 values ranging between 10 and
80 μM); but C. parapsilosisa ATCC 34136 resistant up to
≥ 80 μM of commercial mixture and each the purified
components
Susceptible fungi at 700 ppm with an inhibition halo of
30 mm or more: A. clavatusa, F. oxysporuma,
F. moniliformea, G. candiduma, and P. digitatuma.
Resistant fungi at 700 ppm with an inhibition halo of
22 mm or less: A. nigera, A. flavusa, Rhizopusc sp., and
Mucorc sp
S. sclerotioruma (growth was inhibited around 85% by
5 μM). A. kikuchianaa (ID50 was 10 ppm for growth).
Tilletiab ssp., Helminthosporiuma, Fusariuma ssp., Septoriaa,
Ustilagob, and P. italicuma.
C. gloeosporioidesa (IC50 of the growth: 61.3 μM),
C. lindemuthianuma (IC50 of the growth: 44.8 μM),
F. oxysporuma (IC50 of the growth: 70.7 μM), F. solania
(IC50 of the growth: 61.6 μM), T. rubruma (IC50 of the
growth: 52.06 μM; MIC7: 125 μM; MIC21: 125 μM), and
T. mentagrophytesa (IC50 of the growth: 58.15 μM; MIC7:
125 μM; MIC21: 125 μM)
C. gloeosporioidesa (IC50 of the growth: 168.2 μM),
C. lindemuthianuma (IC50 of the growth: 80.3 μM),
F. oxysporuma, F. solania (IC50 of the growth: 21.6 μM),
T. rubruma (IC50 of the growth: 32.39 μM; MIC7:
62.5 μM; MIC21: 125 μM), and T. mentagrophytesa (IC50
of the growth: 57.85 μM; MIC7: 125 μM; MIC21:
125 μM).
A. nigera ATCC 14604 (MIC of 32 ppm), A. pullulansa
ATCC 9348 (MIC of 16 ppm); G. virensa ATCC 4645
(MIC of 32 ppm), C. globosuma ATCC 6205 (MIC of
16 ppm), P. chrysogenuma ATCC 9480 (MIC of
128 ppm), P. funiculosuma CECT 2914 (MIC of 16 ppm),
C. albicansa ATCC 10231 (MIC of 16 ppm), R. rubrab
CECT 1158 (MIC of 16 ppm), and S. cerevisiaea ATCC
9763 (MIC of 32 ppm).
C. albicansa ATCC 10231 (MIC of 64 ppm), A. nigera
ATCC 46604 (MIC of 125 ppm)
C. albicansa ATCC 10231 (MIC of 125–250 ppm), A. nigera
ATCC 46604 (MIC of 250/500–1000 ppm)
C. albicansa ATCC 10231 (MIC of 3.9 ppm)
A. fumigatusa ATCC 10894 ~ 3.5 log reduction at 50 μg/mL,
C. albicansa ATCC 10231 ~ 2.5 log reduction at
25 μg/mL (starting cell density 107 CFU/mL)
Reference
Gerez et al. 2010
Dreassi et al. 2007
Maldonado et al. 2005
Yagura et al. 1984;
Maiale et al. 2008
Atanasov et al. 2016
Fait et al. 2018
Fait et al. 2018
Kanazawa et al. 1995
Singare and Mhatre
2012
Singare and Mhatre
2012
Singare and Mhatre
2012
Codling et al. 2003
Appl Microbiol Biotechnol
Table 2 (continued)
Surfactant*
Fungal taxa and Oomycota tested/susceptibility**
Reference
3,3′-(2,7-Dioxaoctane) bis(1-decylpyridinium
bromide) [Gemini-QAC 3DOBP-4,10]
S. cerevisiaea NBRC 10217 (CRIC ≥ 0.4 μM; MFC 6.0 μM)
Shirai et al. 2009
C. albicansa 32, 24, and 22 mm/mg of IZD for C10BT,
C12BT, and C16BT, respectively; P. chrysogenuma 23, 19,
and 22 mm/mg of IZD for C10BT, C12BT, and C16BT
respectively (isolates obtained from the operation development Center, Egyptian Petroleum Research Institute,
Egypt)
S. cerevisiaea MIC > 1200 μM for TMEAL-6 Br, 800 μM
for TMEAL-8 Br, 320 μM for TMEAL-10 Br, 80 μM for
TMEAL-12 Br, 500 μM for TMEAL-12 Cl, and 240 μM
for TMEAL-14 Br
C. albicansa MIC > 1200 μM for TMEAL-6 Br, 240 μM for
TMEAL-8 Br, 160 μM for TMEAL-10 Br, 80 μM for
TMEAL-12 Br, 800 μM for TMEAL-12 Cl, and 500 μM
for TMEAL-14 Br
Shaban et al. 2014
N-(3-(butylideneamino)propyl)-N,N-dimethylalky-1-ammonium bromide [C10BT, C12BT, and
C16BT]
N,N′-bis(1-alkyloxy-1-oxopronan-2-yl)-N,N,N′,N
′-tetramethylethane-1,3-diammonium dihalide
[n = alkyl chain length; TMEAL-n X, n = 6, 8, 10,
12, 14, and X = Cl or Br]
Obłąk et al. 2015
R. mucilaginosab MIC > 1200 μM for TMEAL-6 Br, 80 μM
for TMEAL-8 Br, 40 μM for TMEAL-10 Br, 40 μM for
TMEAL-12 Br, 10 μM for TMEAL-12 Cl, and 80 μM for
TMEAL-14 Br
N,N′-bis(1-alkylloxy-1-Oxopronan-2-yl) N,N,N′,N
′-tetramethylpropane-1,2-diammonium dihalide
[n = alkyl chain length; TMPAL-n X, n = 10, 12 and
X = Cl or Br]
S. cerevisiaea CIM 40 μM for TMPAL-10 Br, 500 μM for
TMPAL-12 Br, and 500 μM for TMPAL-12 Cl
C. albicansa CIM 80 μM for TMPAL-10 Br, 500 μM for
TMPAL-12 Br, and 800 μM for TMPAL-12 Cl
Obłąk et al. 2015
R. mucilaginosab CIM 10 μM for TMPAL-10 Br, 10 μM for
TMPAL-12 Br, and 10 μM for TMPAL-12 Cl
N-(3-(benzylideneamino)propyl)-N,N-dimethylalkyl-1-ammonium bromide [n = alkyl chain length;
n = 10 I, n = 12 II, and n = 16 III]
Oleylphosphocholine (OlPC)
Morpholinium chlorides
C. albicansa 25, 31, and 23 mm/mg of IZD for I, II, and III,
respectively; P. chrysogenuma 18, 21, and 17 mm/mg of
IZD for I, II, and III, respectively [II (C12) > I (C10) > III
(C16)] (isolates obtained from the operation development
Center, Egyptian Petroleum Research Institute, Egypt)
C. albicansa MIC50 from 1to 4 mg/L; MFC from 2 to 4 mg/L
(14 strains and clinical isolates obtained from urinary
catheters, blood culture, and disseminated candidiasis
tested)
C. albicansa ATTC 10231 and T. menthagrophytesa ATCC
9533 MFC from 10 to 10,000 mg/mL
Aiad et al. 2014b
Holtappels et al. 2017
Brycki et al. 2010
*Abbreviations commonly found in literature are listed between square brackets
**IC50, concentration value that causes 50% inhibition of growth relative to the control cultures; ID50 (50% inhibitory dose), inhibitor concentration that
produces 50% inhibition; MIC (minimal inhibition concentration), the lowest concentration that exhibited a 100% reduction in growth when compared
with the control cultures; MIC7, the lowest surfactant concentration able to completely inhibit fungal growth after 7 days of exposure to the surfactant;
MIC21, the lowest surfactant concentration able to completely inhibit fungal growth after 21 days of exposure to the surfactant; MIC50, the lowest
concentration of an agent resulting in a growth reduction of ≥ 50% compared to the growth of the control; MIC90, an estimate of the concentration that
inhibits 90% of turbidity compared with that of the control cultures; CRIC, critical respiratory inhibition concentration; MFC, minimal fungicide
concentration; IZD, inhibition zone diameter
a
Phylum Ascomycota
b
Phylum Basidiomycota
c
Phylum Mucoromycota
d
Phylum Oomycota (Kingdom Straminipila)
administration resulted in a significant inhibition of
C. albicans biofilms after 9 days of treatment.
In another study, Obłąk et al. (2015) conducted similar
experiments testing adhesion, biofilm development, and disruption of preformed biofilms of C. albicans and Rhodotorula
mucilaginosa induced by the alanine-derived gemini quaternary ammonium bromides TMPAL-10 Br and TMEAL-12 Br.
The deposition of gemini QACs on polystyrene plates
inhibited the adhesion of R. mucilaginosa cells, with, of the
two, TMPAL-10 Br exhibiting a stronger antiadhesive effect
Appl Microbiol Biotechnol
that caused a higher proportion of killed cells, as evidenced by
fluorescence microscopy and a LIVE/DEAD Viability Kit™.
With respect to the disruption of R. mucilaginosa and
C. albicans biofilms on polystyrene microplates, the two gemini QACs tested were efficient, with TMEAL-12 Br manifesting a slightly stronger effect. Finally, both gemini QACs exhibited antiadhesive properties upon investigation of
C. albicans adhesion to silicone catheters. Only TMEAL-12
Br, however, was able to reduce previously formed biofilms
on those surfaces.
Within the same context, several patents have described the
use of cationic surfactants in the removal of biofilms in industrial systems and in lines and tubing through penetration and
dispersal (Hollis et al. 1995; Labib and Lai 2000; Baldridge
and Michalow 2004). In addition to the disinfectant properties
of cationic surfactants, those compounds—and particularly
the quaternary amines—exhibit a strong interaction with cell
wall constituents of the microorganisms present in biofilms
and thus can facilitate the solubilization of the bacterial and
fungal matter in the biofilm. Furthermore, these surfactants
provide a certain minimal foaming action to an aqueous
cleaning solution that helps to provide a turbulent flow in
the tubing to be cleaned as well as aiding in loosening the
biofilm or debris from the tubing surface.
Bullseye: the plasma membrane
In view of the negative net charge of bacterial and fungal
cellular surfaces, that many antimicrobial agents are cationic
and have a high binding affinity to microbial cells is hardly
surprising. In the example of the cationic surfactants, the presence of a strong positive charge together with a hydrophobic
region is more than enough of a characteristic to enable an
interaction with the cell surface and facilitate an integration
into the cytoplasmic membrane (Gilbert and Moore 2005).
The source of the cationic charge can be variable, but in many
instances, that property can be attributed mainly to the presence of ammonium—i.e., CTAB or guanidinium groups—as
in the example of Nα-lauroyl-L-arginine and dodine, whose
structures are illustrated in Fig. 1.
Guanidine moieties are known to form complexes with
phospholipid head groups by bidentate hydrogen-bonded ion
pairing. This affinity for phospholipids is thought to be responsible for the interaction of those residues with cell membranes and the resulting cell penetration ability and consequent antimicrobial activity (Palermo and Kuroda 2010).
Different mechanisms have been found to be involved in
the antifungal activity of the cationic drugs commonly
employed for the treatment of fungal infections. In general,
the main targets are cell wall polymers—e.g., glucans, chitin,
mannoproteins—, the cell membrane, ergosterol, DNA, the
protein synthesis machinery—topoisomerases, nucleases,
elongation factors, and myristoylation—, and the signal transduction pathways—e.g., protein kinases and protein phosphatases (Sant et al. 2016). Table 3 summarizes the putative
mechanisms described in the literature for the cationic surfactants commonly used as antifungal agents.
An elucidation of the role of lipids in pathogenesis and
target identification for improved therapeutics was pursued
by researchers mainly during the last few years (Sant et al.
2016). A targeting of the fungal cell membrane, the conventional approach, has been extensively explored for the development of antifungal agents. Apart from containing ergosterol, the fungal cell membrane is also rich in
glycerophospholipids and sphingolipids, which components
play an essential role in cellular functions and signal transduction pathways (Dupont et al. 2012). Whereas
glycerophospholipids are composed of glycerol-3-phosphate
containing two fatty-acyl chains along with various polar substituents—like choline, serine, and ethanolamine—,
sphingolipids have a backbone of N-acylated
phytosphingosine or ceramide. Phospholipids such as phosphatidic acid, phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylserine, phosphatidylglycerol, phosphatidylinositol, and cardiolipin along with sphingolipids
such as inositol-phosphate ceramide,
mannosylinositolphosphate ceramide, and
mannosyldiinositolphosphate ceramide are reported as cell
membrane constituents in Saccharomyces cerevisiae (van
der Rest et al. 1995). In studies comparing the phospholipid
and sterol composition of the plasma membrane of
fluconazole-resistant clinical Candida albicans isolates to that
of fluconazole-sensitive ones, Löffler et al. (2000) revealed no
differences in the phospholipid and sterol composition in most
of the strains tested (Löffler et al. 2000), though one resistant
Fig. 1 Chemical structure of a Nα-lauroyl-L-arginine and b dodine. The guanidinium group is indicated in red
Appl Microbiol Biotechnol
strain manifested a decreased amount of ergosterol and had a
lower PC/PE ratio than that of the sensitive strains. These
authors suggested that those changes in the plasma membrane
lipid and sterol composition might be responsible for an altered uptake of drugs and thus for a reduced intracellular accumulation of fluconazole, thereby providing a mechanism
for that azole resistance.
Amphiphiles are known to interact with lipid membranes
so as to affect their structure. For example, natural amphiphiles can be integrated into lipid bilayers or can completely
destroy the bilayer structure to form mixed lipid-surfactant
micelles depending on the amphiphiles’ critical micelle
concentration. In the particular example of CTAB, the
Table 3
critical phenomenon responsible for the antifungal effect
of that compound was suggested to be the reversal of the
cell surface charge from negative to positive without
disrupting the cell membrane, thus enabling the surfactant
to penetrate the cell wall (Vieira and Carmona-Ribeiro
2006). In this regard, Shirai et al. (2009) described how
another family of cationic surfactants, the gemini QACs,
was able to penetrate the cell wall and membrane of
S. cerevisiae, inhibit respiratory enzymes localized in the
mitochondria, and/or destroy organelle membranes. This
evidence led to the assumption that the gemini QAC surfactants produced changes in the permeability of the cytoplasmic membrane in order to reach the interior of the cell.
Reported mechanisms of action of antifungal cationic surfactants
Surfactant
Putative mechanism of action
References
bis(Args)
Cell membrane with increased permeability
Pérez et al. 1996; Tyagi and Tyagi 2014
Bz-Arg-NHCn
Permeabilization and direct damage of the plasma membrane
Induction of oxidative stress
Membrane solubilization with cell lysis at high concentrations
Increase in membrane-surface folding and cell shrinkage, affecting the apparent capacitance of the plasma membrane
Reduction of conductivity in cytoplasm and vacuole interior
Change of cell-surface charge from negative to positive
Protein-function alteration
Interference of cell functions (multisite activity)
Inhibition of respiration of glucose and acetate and active
transport of phosphorus and carbon
Sporulation and conidial-germination inhibition
Alteration of membrane permeability
Inhibition of respiratory function (inhibition of respiratory
enzymes and/or destruction of organelle membranes)
Inhibition of lipid biosynthesis and oxygen uptake
Interference with membrane structure
Disturbance of membrane function
Membrane destabilization and increased permeability
Membrane solubilization with cell lysis
Interference with membrane structure
Loss of membrane potential
Cell permeability alteration
Leakage of cellular constituents
Inhibition of sterol biosynthesis (inhibition of ∆8 → ∆7
isomerase), causing the accumulation of fecosterol and
ergosta-8-en-3ß-ol in Ustilago maydis and Saccharomyces
cerevisiae and fecosterol and
ergosterol-8,22,24(27)-trien-3ß-ol in Botrytis cinerea and
Penicillium expansum
Membrane damage
Fait et al. 2018
CTAB
Dodine
3DOBP-4,10
Guazatine
LAE
Meltatox
MAPD
QACs
Raicu 1998; Gupta et al. 2002; Vieira and Carmona-Ribeiro
2006; Posadas et al. 2012; Sandle et al. 2014; Mahmoud
2016; Fait et al. 2018
Szkolnik and Gilpatrick 1969; Yoder and Klos 1976; Deepak
et al. 2006; Luz et al. 2007; Tóth et al. 2012; Posadas et al.
2012
Shirai et al. 2009
Yagura et al. 1984; Maldonado et al. 2005; Dreassi et al.
2007; Maiale et al. 2008; Gerez et al. 2010; Atanasov et al.
2016
Kanazawa et al. 1995
Benyagoub and Bélanger 1995
Codling et al. 2003
Alteration of membrane permeability
Hegstad et al. 2010
Leakage of cellular constituents
At high concentrations, can solubilize hydrophobic cell
membrane components by forming mixed micellar aggregates
Disruption and denaturation of structural proteins and enzymes
Appl Microbiol Biotechnol
Model membrane systems for the study
of the antifungal mechanism of amphiphilic
compounds
A study of the interaction of bioactive compounds with
biomembranes—a complex phenomenon from both the
chemical and the physicochemical points of view—may provide fundamental information about the mechanism involved,
thus expanding our knowledge and offering an opportunity to
identify further potential therapeutic targets. Within this context, and in consideration of the role of the plasma membrane,
experiments involving different types of model membranes—
such as lipid mono- or bilayers or lipid vesicles (liposomes)—
are therefore of paramount necessity in guiding the development of new antifungal agents.
Membrane models differ in their complexity (e.g., monolayers, supported bilayers, or vesicles) as well as in their lipidic nature (e.g., saturated or unsaturated lipids, zwitterions, or
charged lipids) and their physical state (involving the fluidity
or rigidity of the acyl chains). Nevertheless, a simple lipid
mixture composed of 75 mol% 1-palmitoyl-2-oleoyl-snglycero-3-phosphocholine and 25 mol% 1-palmitoyl-2oleoyl-sn-glycero-3-phospho-l-serine has been found to
match the fluidity and charge of the phospholipid composition
in the ascomycete (example in point, C. albicans) cellular
membrane (Stenbæk et al. 2017).
Valuable information about antifungal mechanisms based
on membrane interactions can be obtained through membrane
models along with other biophysical techniques. GonzálezJaramillo et al. (2017) have performed a detailed biophysical
study on the interaction of the biosurfactant fengycin C with
model dipalmitoylphosphatidylcholine (DPPC) membranes
(González-Jaramillo et al. 2017). Combining differential scanning calorimetry and fluorescence polarization probe measurements with Fourier transform infrared spectroscopy, those
authors demonstrated that fengycin C alters the thermotropic
phase transitions of DPPC and is laterally segregated in the
fluid bilayer-forming domains, without affecting the hydrophobic interior of the membrane. Fengycin-rich domains,
where the surrounding DPPC molecules are highly
Fig. 2 Antifungal mechanism by of Bz-Arg-NHC n . a Control
Colletotrichum spp. conidia. b Membrane permeabilization (evidenced
by propidium iodide uptake) induced in conidia of C. lindemuthianum by
exposure to 400 μM Bz-Arg-NHC12. c Detection of reactive oxygen
dehydrated, may well constitute sites of membrane permeabilization leading to a leaky target membrane.
Studies on the plausible mode of action of cationic surfactants support a membranolytic or detergent-like effect similar
to that of many membrane-active antimicrobial peptides that
should make the development of a complete resistance difficult for the microorganisms (Bahar and Ren 2013).
The example of the Bz-Arg-NHCn family
of arginine-based surfactants
In the particular example of arginine-based surfactants, our
own previous work evidenced that, according to the relationship between the chemical structure and the biologic activity
of the Bz-Arg-NHCn, the shortness of the alkyl chain was
correlated with the strength of the fungistatic activity, whereas, for the bactericidal and/or bacteriostatic capability, the
length the alkyl chain correlated with the extensiveness of
the antimicrobial potency (Fait et al. 2015; Fait et al. 2018).
Considering the differences between bacterial and fungal
cells, we suggested that the hydrophobic character of the surfactant molecules might lead to a less efficient internalization,
probably because of a strong interaction of those amphiphiles
with the fungal lipid membrane. According to Castillo et al.
(2006), the difference observed in the behavior of this kind of
surfactant towards bacteria and fungi could be explained on
the basis of an adequate lipophilic-hydrophilic balance of the
molecule, which causes a disruption of plasma membranes or
affects intracellular processes, which actions are described in
the previous section (Castillo et al. 2006). This hypothesis
could explain the higher fungicidal effect of Bz-Arg-NHC10
than Bz-Arg-NHC12 for almost all the fungal species tested so
far.
The analysis of fungal membrane integrity and the qualitative production of reactive oxygen species (Fig. 2) suggested
both membrane permeabilization and the induction of oxidative stress to be a part of the antifungal mechanism involved in
the interruption of normal conidial development by Bz-ArgNHCn (Fait et al. 2018).
species (evidenced by the uptake of the substrate 3,3-diaminobenzidine)
produced in the conidia of C. gloeosporioides through the exposure to
400 μM Bz-Arg-NHC12. Bars: 30 μm (for more details see Fait et al.
2018)
Appl Microbiol Biotechnol
On the basis of the internalization of surfactant molecules as part of the antifungal mechanism and the presence of the arginine moiety in these surfactant species,
different models can be proposed to explain their translocation into the cytoplasm through the membrane: (i) direct
uptake, which internalization would involve destabilization of the membrane in an energy- and temperatureindependent manner; (ii) inverted micelle formation; (iii)
pore formation; and/or (iv) electroporation-like permeabilization (Bechara and Sagan 2013). In view of the molecular structure of Bz-Arg-NHCn, the truncated conical
shape of these molecules supports the hypothesis that
deals with the formation of transient pores as the main
mechanism. In general, facilitation of pore formation by
molecules with a positive spontaneous curvature (such as
lysophospholipids) is explained by a decrease in the free
energy per unit length—i.e., the line tension—that is required to form the edges of a pore whose overall geometric monolayer curvature is positive. In contrast,
nonlamellar lipids with a negative intrinsic curvature
(such as PE) increase that free energy of pore creation.
In a toroid, a positive curvature is found perpendicular to
the plane of the membrane, whereas a negative curvature
is present in that plane all around the pore. Considering
that (i) PE—with an inverted conical shape—may adopt
different structures in the membrane, i.e., lamellar or
inverted micelles, (ii) strong head group interactions will
be established through the formation of H bonds, and that
(iii) fungal membranes are rich in PE, a transient pore
formation—through which the surfactant molecules can
translocate and/or diffuse and reach the cytoplasm—
Table 4 Common uses of
cationic surfactants for fungal
control
Surfactant
CTAB
Meltatox
Dodine
Guazatine
Iminoctadine
LAE
MAPD
Fig. 3 Transient pore formation as the proposed model for the effect of BzArg-NHCn surfactants on fungal membranes. On the basis of fungal
membrane’s composition—rich in phosphatidylethanolamine—and
because strong head group interactions could be established through the
formation of H bonds between the surfactant head group and the
hydrophilic portion of phospholipids, transient pore formation could
facilitate the translocation and/or diffusion of surfactant molecules into the
cell cytoplasm. PC phosphatidylcholine, PE phosphatidylethanolamine, S
Bz-Arg-NHCn
would be the most appropriate mechanistic model for
explaining the antifungal effect of these surfactants
(Fig. 3). Nevertheless, in order to confirm our hypothesis,
further trials with model membrane systems still are
needed.
Uses of cationic surfactants as antifungal
agents
Cationic surfactants as antifungal agents can be considered as
multisite-active compounds since those amphipaths can exert
their antifungal activity through different modes of action.
Indeed, such detergents can accordingly be used as fungicides
Uses
Chemical agent with fungicide or fungistatic activity
Potential use in treatment of mycotic corneal ulcers
Systemic fungicide used in agriculture
Fungicide with preventive action and curative effect especially indicated for the control of
scabies in apple and pear. The surfactant also has an eradicating effect when applied 30 to
36 h after the infection was produced. It is used to control scabies in pecan nuts; leaf spot on
cherries, olive, redcurrants, celery, and other crops as well as leaf diseases in strawberry and
can be used for other fruit plants, ornamental or otherwise. The cationic tenside type
dodemorph isomers and dodine as well as the phosphonic acid salts were also well tolerated
by pearl millet germlings. Multisite contact activity
The surfactant is widely used in agriculture to control a wide range of seed-borne diseases of
cereals. On citrus fruit, it is used as a bulk dip after harvest and in the packing line as a spray.
A commercial product, it is a nonsystemic-contact fungicide with potent anti-Candida
activity, superior to that of the commercial fluconazole, and is a polyamine-oxidase inhibitor
in plants with a Ki ~ 10−8 M.
Protectant. Control of fungal pathogens on citrus, ornamental and fruit trees, lawns and turf.
Usually formulated as a seed dressing
Preservative for the food industry. Commercialized as Mirenat™
Contact lens multipurpose disinfecting solution, commercial name Opti-Free Express™ (Alcon)
Appl Microbiol Biotechnol
and/or as fungistatic agents. Depending on where in the disease cycle or deterioration process those cationic surfactants
act (Balba 2007), they can play a role as (i) protectants
(through contact) or preventive fungicides that are effective
before colonization, such as in the example of dodine and
iminoctadine; (ii) curative agents that are effective against
the fungus growing in the host tissue or the fungal product
deposited after the occurrence of the infecting spores’ germination and therefore have curative properties; and (iii)
eradicants or antisporulant fungicides that are capable of stopping sporulation by the organism, as reported for dodine.
These compounds can furthermore be used in the field of
agriculture to control postharvest diseases as well as in human
and veterinary medicine as antifungal agents in topical formulations against opportunistic mycoses in addition to being disinfectants. Table 4 summarizes common uses of cationic surfactants as antifungal agents.
Perspectives
At the present, amino acid-based cationic surfactants are recognized not only as potential antifungal agents, but also as
promising enhancers and solubilizers of the already existing
antifungal drugs, by that means contributing to a widening of
the therapeutic window of those pharmacons. Likewise, the
use of these compounds for the pretreatment of surfaces represents a promising alternative to the quaternary ammonium
salts as biocidal agents with biomedical applications, focusing
on the control of the colonization of those sites by pathogenic
microorganisms and the subsequent formation of biofilms.
Because of all the advantages cited in this review, the
arginine-based cationic surfactants deserve the attention of
future research in the field of biochemistry and the biomedical
sciences as well as in the realm of agriculture and the pharmaceutical and food industries.
Acknowledgments MEF was awarded a CONICET fellowship. SRM,
MCNS, and GLG are members of CONICET. LB is member of the
CICPBA as a career investigator. Dr. Donald F. Haggerty, a retired academic career investigator and native English speaker, edited the final
version of the manuscript.
Funding information This study was funded by MINCyT (PICT 20132531 and PICT 2015-1620), CAPES-MINCyT (017/2014), and UNLP
(X11-682).
Compliance with ethical standards
This article does not contain any studies with human participants or animals performed by any of the authors.
Conflict of interest The authors declare that they have no conflict of
interest.
References
Acmite Mark (2016) Global surfactant market. Market report. In: Acmite
Mark. Intell. http://www.acmite.com/brochure/Brochure-GlobalSurfactant-Market-Report.pdf. Accessed 6 Sept 2018
Aiad I, El-Sukkary MM, Soliman EA, El-Awady MY, Shaban SM
(2014a) Inhibition of mild steel corrosion in acidic medium by some
cationic surfactants. J Ind Eng Chem 20:3524–3535. https://doi.org/
10.1016/j.jiec.2013.12.045
Aiad I, El-Sukkary MM, Soliman EA, El-Awady MY, Shaban SM
(2014b) Characterization, surface properties and biological activity
of new prepared cationic surfactants. J Ind Eng Chem 20:1633–
1640. https://doi.org/10.1016/j.jiec.2013.08.010
Anderson SE, Shane H, Long C, Lukomska E, Meade BJ, Marshall NB
(2016) Evaluation of the irritancy and hypersensitivity potential following topical application of didecyldimethylammonium chloride. J
Immunotoxicol 13:557–566. https://doi.org/10.3109/1547691X.
2016.1140854
Atanasov KE, Barboza-Barquero L, Tiburcio AF, Alcázar R (2016)
Genome wide association mapping for the tolerance to the polyamine oxidase inhibitor guazatine in Arabidopsis thaliana. Front
Plant Sci 7:1–11. https://doi.org/10.3389/fpls.2016.00401
Bahar AA, Ren D (2013) Antimicrobial peptides. Pharmaceuticals 6:
1543–1575. https://doi.org/10.3390/ph6121543
Balba H (2007) Review of strobilurin fungicide chemicals. J Environ Sci
Health B 42:441–451. https://doi.org/10.1080/03601230701316465
Baldridge JW, Michalow A (2004) Biofilm reduction in crossflow filtration systems. US Grant US6699391B2. 1–7
Bechara C, Sagan S (2013) Cell-penetrating peptides: 20 years later,
where do we stand? FEBS Lett 587:1693–1702. https://doi.org/10.
1016/j.febslet.2013.04.031
Benyagoub M, Bélanger RR (1995) Development of a mutant strain of
Sporothrix flocculosa with resistance to dodemorph-acetate.
Phytopathology 85:766–770. https://doi.org/10.1094/Phyto-85-766
Bordes R, Holmberg K (2015) Amino acid-based surfactants—do they
deserve more attention? Adv Colloid Interf Sci 222:79–91. https://
doi.org/10.1016/j.cis.2014.10.013
Brycki B, Dega-Szafran Z, Mirska I (2010) Synthesis and antimicrobial
activities of some quaternary morpholinium chlorides. Polish J
Microbiol 59:49–53
Bseiso E, Nasr M, Abd El Gawad N, Sammour O (2015) Recent advances in topical formulation carriers of antifungal agents. Indian J
Dermatol Venereol Leprol 81:457. https://doi.org/10.4103/03786323.162328
Castillo JA (2006) Comparative study of the antimicrobial activity of
bis(N-caproyl-L-arginine)-1,3-propanediamine dihydrochloride
and chlorhexidine dihydrochloride against Staphylococcus aureus
and Escherichia coli. J Antimicrob Chemother 57:691–698.
https://doi.org/10.1093/jac/dkl012
Castillo JA, Infante MR, Manresa A, Vinardell MP, Mitjans M, Clapés P
(2006) Chemoenzymatic synthesis and antimicrobial and
haemolytic activities of amphiphilic bis(phenylacetylarginine) derivatives. ChemMedChem 1:1091–1098. https://doi.org/10.1002/
cmdc.200600148
Chandra N, Tyagi VK (2013) Synthesis, properties, and applications of
amino acids based surfactants: a review. J Dispers Sci Technol 34:
800–808. https://doi.org/10.1080/01932691.2012.695967
Chen Y, Geurts M, Sjollema SB, Kramer NI, Hermens JLM, Droge STJ
(2014) Acute toxicity of the cationic surfactant C12-benzalkonium
in different bioassays: how test design affects bioavailability and
effect concentrations. Environ Toxicol Chem 33:606–615. https://
doi.org/10.1002/etc.2465
Codling CE, Maillard JY, Russell AD (2003) Aspects of the antimicrobial
mechanisms of action of a polyquaternium and an amidoamine. J
Appl Microbiol Biotechnol
Antimicrob Chemother 51:1153–1158. https://doi.org/10.1093/jac/
dkg228
Colomer A, Pinazo A, Manresa MA, Vinardell MP, Mitjans M, Infante
MR, Pérez L (2011) Cationic surfactants derived from lysine: effects
of their structure and charge type on antimicrobial and hemolytic
activities. J Med Chem 54:989–1002. https://doi.org/10.1021/
jm101315k
Dancer SJ (2009) The role of environmental cleaning in the control of
hospital-acquired infection. J Hosp Infect 73:378–385. https://doi.
org/10.1016/j.jhin.2009.03.030
Deepak SA, Basavaraju P, Chaluvaraju G, Shetty NP, Oros G, Shekar H
(2006) Developmental stage response of pearl millet downy mildew
(Sclerospora graminicola) to fungicides. Appl Ecol Environ Res 4:
125–149. https://doi.org/10.15666/aeer/0402_125149
Desai JV, Mitchell AP, Andes DR (2014) Fungal biofilms, drug resistance, and recurrent infection. Cold Spring Harb Perspect Med 4:
a019729. https://doi.org/10.1101/cshperspect.a019729
Di Nica V, Gallet J, Villa S, Mezzanotte V (2017) Toxicity of quaternary
ammonium compounds (QACs) as single compounds and mixtures
to aquatic non-target microorganisms: experimental data and predictive models. Ecotoxicol Environ Saf 142:567–577. https://doi.org/
10.1016/j.ecoenv.2017.04.028
Dreassi E, Zizzari AT, D’Arezzo S, Visca P, Botta M (2007) Analysis of
guazatine mixture by LC and LC–MS and antimycotic activity determination of principal components. J Pharm Biomed Anal 43:
1499–1506. https://doi.org/10.1016/j.jpba.2006.10.029
Dupont S, Lemetais G, Ferreira T, Cayot P, Gervais P, Beney L (2012)
Ergosterol biosynthesis: a fungal pathway for life on land?
Evolution (N Y):1–8. https://doi.org/10.5061/dryad.pd28pm7n
Dusane DH, Dam S, Nancharaiah YV, Kumar A, Venugopalan VP,
Zinjarde SS (2012) Disruption of Yarrowia lipolytica biofilms by
rhamnolipid biosurfactant. Aquat Biosyst 8:17. https://doi.org/10.
1186/2046-9063-8-17
Fait ME, Garrote GL, Clapés P, Tanco S, Lorenzo J, Morcelle SR (2015)
Biocatalytic synthesis, antimicrobial properties and toxicity studies
of arginine derivative surfactants. Amino Acids 47:1465–1477.
https://doi.org/10.1007/s00726-015-1979-0
Fait ME, da Costa H PS, Freitas C DT, Bakás L, Morcelle SR (2018)
Antifungal activity of arginine-based surfactants. Curr Bioact
Compd 14:1–9. https://doi.org/10.2174/
1573407214666180131161302
Florence AT, Atwood D (2006) Surfactants. In: Physicochemical principles of pharmacy, 4th edn. Pharmaceutical Press, London
Gaikwad KK, Lee SM, Lee JS, Lee YS (2017) Development of antimicrobial polyolefin films containing lauroyl arginate and their use in
the packaging of strawberries. J Food Meas Charact 11:1706–1716.
https://doi.org/10.1007/s11694-017-9551-0
Gecol H, Ergican E, Fuchs A (2004) Molecular level separation of arsenic
(V) from water using cationic surfactant micelles and ultrafiltration
membrane. J Memb Sci 241:105–119. https://doi.org/10.1016/j.
memsci.2004.04.026
Gerez CL, Carbajo MS, Rollán G, Torres Leal G, Font de Valdez G
(2010) Inhibition of citrus fungal pathogens by using lactic acid
bacteria. J Food Sci 75:M354–M359. https://doi.org/10.1111/j.
1750-3841.2010.01671.x
Gerova M, Rodrigues F, Lamère J-F, Dobrev A, Fery-Forgues S (2008)
Self-assembly properties of some chiral N-palmitoyl amino acid
surfactants in aqueous solution. J Colloid Interface Sci 319:526–
533. https://doi.org/10.1016/j.jcis.2007.12.004
Gilbert P, Moore LE (2005) Cationic antiseptics: diversity of action under
a common epithet. J Appl Microbiol 99:703–715. https://doi.org/10.
1111/j.1365-2672.2005.02664.x
Gong T, Zhang X, Li Y, Xian Q (2016) Formation and toxicity of halogenated disinfection byproducts resulting from linear alkylbenzene
sulfonates. Chemosphere 149:70–75. https://doi.org/10.1016/j.
chemosphere.2016.01.067
González-Jaramillo LM, Aranda FJ, Teruel JA, Villegas-Escobar V, Ortiz
A (2017) Antimycotic activity of fengycin C biosurfactant and its
interaction with phosphatidylcholine model membranes. Colloids
Surf B Biointerfaces 156:114–122. https://doi.org/10.1016/j.
colsurfb.2017.05.021
Grand View Research (2016) Surfactants market revenue is expected to
grow at CAGR of 5.4% owing to enhanced demand in personal care
industry till 2022: Grand View Research, Inc. In: Gd. View Res.
http://globenewswire.com/news-release/2016/02/11/809799/0/en/
Surfactants-Market-Revenue-Is-Expected-To-Grow-At-CAGR-Of5-4-Owing-To-Enhanced-Demand-In-Personal-Care-Industry-Till2022-Grand-View-Research-Inc.html. Accessed 2 Sept 2018
Gupta AK, Ahmad I, Summerbell RC (2002) Fungicidal activities of
commonly used disinfectants and antifungal pharmaceutical spray
preparations against clinical strains of Aspergillus and Candida species. Med Mycol 40:201–208. https://doi.org/10.1080/mmy.40.2.
201.208
Hanson JR (2008) Chemistry of fungi, RSC Publis. Royal Society of
Chemistry, Cambridge
Hawser SP, Douglas LJ (1994) Biofilm formation by Candida species on
the surface of catheter materials in vitro. Infect Immun 62:915–921
Hay R (2017) Superficial fungal infections. Med (United Kingdom) 45:
707–710. https://doi.org/10.1016/j.mpmed.2017.08.006
Hayes DG (2009) Surfactants overview and industrial state-of-the-art. In:
Hayes DG, Kitamoto D, Solaiman D, Ashby RD (eds) Biobased
surfactants and detergents: synthesis, properties, and applications.
AOCS Press, Illinois, pp 3–28
He K, Xu L (2017) Unique mixtures of anionic/cationic surfactants: a
new approach to enhance surfactant performance in liquids-rich
shale reservoirs. SPE Prod Oper 33:1–8. https://doi.org/10.2118/
184515-PA
Hegazy MA, El-Etre AY, El-Shafaie M, Berry KM (2016) Novel cationic
surfactants for corrosion inhibition of carbon steel pipelines in oil
and gas wells applications. J Mol Liq 214:347–356. https://doi.org/
10.1016/j.molliq.2015.11.047
Hegstad K, Langsrud S, Lunestad BT, Scheie AA, Sunde M, Yazdankhah
SP (2010) Does the wide use of quaternary ammonium compounds
enhance the selection and spread of antimicrobial resistance and thus
threaten our health? Microb Drug Resist 16:91–104. https://doi.org/
10.1089/mdr.2009.0120
Hollis CG, Terry JP, Jaquess PA (1995) Methods for removing biofilm
from or preventing buildup thereof on surfaces in industrial water
systems. 1–10
Holtappels M, Swinnen E, De Groef L, Wuyts J, Moons L, Lagrou K,
Van Dijck P, Kucharíková S (2017) Antifungal activity of
oleylphosphocholine on in vitro and in vitro Candida albicans
biofilms. Antimicrob Agents Chemother 62:e01767–e01717.
https://doi.org/10.1128/AAC.01767-17
Infante MR, Pérez L, Morán C, Pons R, Pinazo A (2009) Synthesis,
aggregation properties, and applications of biosurfactants derived
from arginine. In: Hayes DG, Kitamoto D, Solaiman D, Ashby R
(eds) Biobased surfactants and detergents: synthesis, properties, and
applications. AOCS Press, Illinois, pp 351–387
Isaac J, Scheinman PL (2017) Benzalkonium chloride. Dermatitis 28:
346–352. https://doi.org/10.1097/DER.0000000000000316
Jabra-Rizk MA, Falkler WA, Meiller TF (2004) Fungal biofilms and drug
resistance. Emerg Infect Dis 10:14–19. https://doi.org/10.3201/
eid1001.030119
Jennings MC, Minbiole KPC, Wuest WM (2015) Quaternary ammonium
compounds: an antimicrobial mainstay and platform for innovation
to address bacterial resistance. ACS Infect Dis 1:288–303. https://
doi.org/10.1021/acsinfecdis.5b00047
Jessop PG, Ahmadpour F, Buczynski MA, Burns TJ, Green II NB,
Korwin R, Long D, Massad SK, Manley JB, Omidbakhsh N,
Pearl R, Pereira S, Predale RA, Sliva PG, VanderBilt H, Weller S,
Wolf MH (2015) Opportunities for greener alternatives in chemical
Appl Microbiol Biotechnol
formulations. Green Chem 17:2664–2678. https://doi.org/10.1039/
C4GC02261K
Jiang Q, Yue D, Nie Y, Xu X, He Y, Zhang S, Wagner E, Gu Z (2016)
Specially-made lipid-based assemblies for improving transmembrane gene delivery: comparison of basic amino acid residue rich
periphery. Mol Pharm 13:1809–1821. https://doi.org/10.1021/acs.
molpharmaceut.5b00967
Johansson J, Somasundarau P (eds) (2007) Handbook for cleaning/
decontamination of surfaces. Elsevier, Amsterdam
Kanazawa A, Ikeda T, Endo T (1995) A novel approach to mode of action
of cationic biocides: morphological effect on antibacterial activity. J
Appl Bacteriol 78:55–60. https://doi.org/10.1111/j.1365-2672.1995.
tb01673.x
Labib ME, Lai C-Y (2000) Cleaning method for removing biofilm and
debris from lines and tubing. 1–10
Lavorgna M, Russo C, D’Abrosca B, Parrella A, Isidori M (2016)
Toxicity and genotoxicity of the quaternary ammonium compound
benzalkonium chloride (BAC) using Daphnia magna and
Ceriodaphnia dubia as model systems. Environ Pollut 210:34–39.
https://doi.org/10.1016/j.envpol.2015.11.042
Li Z, Alessi D, Zhang P, Bowman RS (2002) Organo-illite as a low
permeability sorbent to retard migration of anionic contaminants. J
Environ Eng 128:583–587. https://doi.org/10.1061/(ASCE)07339372(2002)128:7(583)
Li Z, Willms CA, Kniola K (2003) Removal of anionic contaminants
using surfactant-modified palygorskite and sepiolite. Clay Clay
Miner 51:445–451. https://doi.org/10.1346/CCMN.2003.0510411
Li Y, Zhang W, Kong B, Puerto M, Bao X, Sha O, Shen Z, Yang Y, Liu Y,
Gu S, Miller C, Hirasaki GJ (2014) Mixtures of anionic-cationic
surfactants: a new approach for enhanced oil recovery in low-salinity, high-temperature sandstone reservoir. In: SPE Improved Oil
Recovery Symposium. Society of Petroleum Engineers, pp 1164–
1177
Löffler J, Einsele H, Hebart H, Schumacher U, Hrastnik C, Daum G
(2000) Phospholipid and sterol analysis of plasma membranes of
azole-resistant Candida albicans strains. FEMS Microbiol Lett
185:59–63. https://doi.org/10.1111/j.1574-6968.2000.tb09040.x
Lozano N, Pérez L, Pons R, Pinazo A (2011) Diacyl glycerol argininebased surfactants: biological and physicochemical properties of
catanionic formulations. Amino Acids 40:721–729. https://doi.org/
10.1007/s00726-010-0710-4
Luz C, Netto MCB, Rocha LFN (2007) In vitro susceptibility to fungicides by invertebrate-pathogenic and saprobic fungi.
Mycopathologia 164:39–47. https://doi.org/10.1007/s11046-0079020-0
Mahmoud YA (2016) Fungal keratitis efficient treatments using surface
active agents (cetrimide): an overview. J Bacteriol Mycol Open
Access 2:3–7. https://doi.org/10.15406/jbmoa.2016.02.00026
Maiale SJ, Marina M, Sánchez DH, Pieckenstain FL, Ruiz OA (2008)
In vitro and in vivo inhibition of plant polyamine oxidase activity by
polyamine analogues. Phytochemistry 69:2552–2558. https://doi.
org/10.1016/j.phytochem.2008.07.003
Maier C, Zeeb B, Weiss J (2014) Investigations into aggregate formation
with oppositely charged oil-in-water emulsions at different pH
values. Colloids Surfaces B Biointerfaces 117:368–375. https://
doi.org/10.1016/j.colsurfb.2014.03.012
Maldonado MC, Santa Runco R, Navarro AR (2005) Isolation, identification and antifungal susceptibility of lemon pathogenic and non
pathogenic fungi. Rev Iberoam Micol 22:57–59. https://doi.org/10.
1016/S1130-1406(05)70009-7
Mallo AC, Nitiu DS, Elíades LA, Saparrat MCN (2017) Fungal degradation of cellulosic materials used as support for cultural heritage. Int J
Conserv Sci 8:619–632
Manrique Y, Gibis M, Schmidt H, Weiss J (2017) Influence of application
sequence and timing of eugenol and lauric arginate (LAE) on
survival of spoilage organisms. Food Microbiol 64:210–218.
https://doi.org/10.1016/j.fm.2017.01.002
Mao X, Jiang R, Xiao W, Yu J (2015) Use of surfactants for the remediation of contaminated soils: a review. J Hazard Mater 285:419–435.
https://doi.org/10.1016/j.jhazmat.2014.12.009
Messier C, Epifano F, Genovese S, Grenier D (2011) Inhibition of
Candida albicans biofilm formation and yeast-hyphal transition by
4-hydroxycordoin. Phytomedicine 18:380–383. https://doi.org/10.
1016/j.phymed.2011.01.013
Morán C, Clapés P, Comelles F, García T, Pérez L, Vinardell P, Mitjans
M, Infante MR (2001) Chemical structure/property relationship in
single-chain arginine surfactants. Langmuir 17:5071–5075. https://
doi.org/10.1021/la010375d
MycoBank (2018) Fungal databases, nomenclature & species banks.
International Mycological Association. http://www.mycobank.org/.
Accessed 15 Sept 2018
Negm NA, Mohamed AS (2008) Synthesis, characterization and biological activity of sugar-based gemini cationic amphiphiles. J
Surfactant Deterg 11:215–221. https://doi.org/10.1007/s11743008-1071-9
Nogueira DR, Scheeren LE, Macedo LB, Marcolino AIP, Pilar Vinardell
M, Mitjans M, Rosa Infante M, Farooqi AA, Rolim CMB (2015)
Inclusion of a pH-responsive amino acid-based amphiphile in
methotrexate-loaded chitosan nanoparticles as a delivery strategy
in cancer therapy. Amino Acids 48:157–168. https://doi.org/10.
1007/s00726-015-2075-1
Obłąk E, Piecuch A, Krasowska A, Łuczyński J (2013) Antifungal activity of gemini quaternary ammonium salts. Microbiol Res 168:630–
638. https://doi.org/10.1016/j.micres.2013.06.001
Obłąk E, Piecuch A, Dworniczek E, Olejniczak T (2015) The influence of
biodegradable gemini surfactants, N,N’-bis(1-decyloxy-1oxopronan-2-yl)-N,N,N’,N’ tetramethylpropane-1,3-diammonium
dibromide and N,N’-bis(1-dodecyloxy-1-oxopronan-2-yl) N,N,N’,
N’-tetramethylethane-1,2-diammonium dibromide. Fungal Biof J
Oleo Sci 64:527–537. https://doi.org/10.5650/jos.ess14195
Occams Business Research & Consulting (2017) Global surfactants market insights, opportunity analysis, market shares and forecast 20172023. In: Data, Mark Anal http://www.occamsresearch.com/
surfactants-market. Accessed 2 Oct 2017
Palermo EF, Kuroda K (2010) Structural determinants of antimicrobial
activity in polymers which mimic host defense peptides. Appl
Microbiol Biotechnol 87:1605–1615. https://doi.org/10.1007/
s00253-010-2687-z
Pang X, Chu C-C (2010) Synthesis, characterization and biodegradation
of functionalized amino acid-based poly(ester amide)s. Biomaterials
31:3745–3754. https://doi.org/10.1016/j.biomaterials.2010.01.027
Peña LC, Argarañá MF, De Zan MM, Giorello A, Antuña S, Prieto CC,
Veaute CMI, Müller DM (2017) New amphiphilic amino acid derivatives for efficient DNA transfection in vitro. Adv Chem Eng Sci
07:191–205. https://doi.org/10.4236/aces.2017.72014
Pérez L, Torres JL, Manresa A, Solans C, Infante MR (1996) Synthesis,
aggregation, and biological properties of a new class of gemini cationic amphiphilic compounds from arginine, bis(Args) †. Langmuir
12:5296–5301. https://doi.org/10.1021/la960301f
Pérez L, Pinazo A, Teresa García M, Lozano M, Manresa A, Angelet M,
Pilar Vinardell M, Mitjans M, Pons R, Rosa Infante M (2009)
Cationic surfactants from lysine: synthesis, micellization and biological evaluation. Eur J Med Chem 44:1884–1892. https://doi.org/
10.1016/j.ejmech.2008.11.003
Perfect JR (2017) The antifungal pipeline: a reality check. Nat Rev Drug
Discov 16:603–616. https://doi.org/10.1038/nrd.2017.46
Pinazo A, Pons R, Pérez L, Infante MR (2011) Amino acids as raw
material for biocompatible surfactants. Ind Eng Chem Res 50:
4805–4817. https://doi.org/10.1021/ie1014348
Pinazo A, Manresa MA, Marques AM, Bustelo M, Espuny MJ, Pérez L
(2016) Amino acid–based surfactants: new antimicrobial agents.
Appl Microbiol Biotechnol
Adv Colloid Interf Sci 228:17–39. https://doi.org/10.1016/j.cis.
2015.11.007
Posadas JB, Comerio RM, Mini JI, Nussenbaum AL, Lecuona RE (2012)
A novel dodine-free selective medium based on the use of cetyl
trimethyl ammonium bromide (CTAB) to isolate Beauveria
bassiana, Metarhizium anisopliae sensu lato and Paecilomyces
lilacinus from soil. Mycologia 104:974–980. https://doi.org/10.
3852/11-234
Raicu V (1998) Effects of cetyltrimethylammonium bromide (CTAB)
surfactant upon the dielectric properties of yeast cells. Biochim
Biophys Acta - Gen Subj 1379:7–15. https://doi.org/10.1016/
S0304-4165(97)00056-1
Ramage G, Saville SP, Wickes BL, Lopez-Ribot JL (2002) Inhibition of
Candida albicans biofilm formation by farnesol, a quorum-sensing
molecule. Appl Environ Microbiol 68:5459–5463. https://doi.org/
10.1128/AEM.68.11.5459-5463.2002
Rosa M, del Carmen Morán M, da Graça Miguel M, Lindman B (2007)
The association of DNA and stable catanionic amino acid-based
vesicles. Colloids Surfaces A Physicochem Eng Asp 301:361–
375. https://doi.org/10.1016/j.colsurfa.2006.12.082
Sandle T, Vijayakumar R, Saleh Al Aboody M, Saravanakumar S (2014)
In vitro fungicidal activity of biocides against pharmaceutical environmental fungal isolates. J Appl Microbiol 117:1267–1273. https://
doi.org/10.1111/jam.12628
Sant DG, Tupe SG, Ramana CV, Deshpande MV (2016) Fungal cell
membrane-promising drug target for antifungal therapy. J Appl
Microbiol 121:1498–1510. https://doi.org/10.1111/jam.13301
Shaban SM, Aiad I, El-Sukkary MM, Soliman EA, El-Awady MY (2014)
Synthesis, surface, thermodynamic properties and biological activity
of dimethylaminopropylamine surfactants. J Ind Eng Chem 20:
4194–4201. https://doi.org/10.1016/j.jiec.2014.01.020
Shaban SM, Aiad I, El-Sukkary MM, Soliman EA, El-Awady MY
(2015a) Evaluation of some cationic surfactants based on
dimethylaminopropylamine as corrosion inhibitors. J Ind Eng
Chem 21:1029–1038. https://doi.org/10.1016/j.jiec.2014.05.012
Shaban SM, Aiad I, Fetouh HA, Maher A (2015b) Amidoamine double
tailed cationic surfactant based on dimethylaminopropylamine: synthesis, characterization and evaluation as biocide. J Mol Liq 212:
699–707. https://doi.org/10.1016/j.molliq.2015.10.024
Shirai A, Sumitomo T, Kurimoto M, Maseda H, Kourai H (2009) The
mode of the antifungal activity of gemini-pyridinium salt against
yeast. Biocontrol Sci 14:13–20. https://doi.org/10.4265/bio.14.13
Singare PU, Mhatre JD (2012) Cationic surfactants from arginine: synthesis and physicochemical properties. Am J Chem 2:186–190.
https://doi.org/10.5923/j.chemistry.20120204.02
Singh A, Tyagi VK (2014) Arginine based novel cationic surfactants: a
review. Tenside Surfactants Deterg 51:202–214. https://doi.org/10.
3139/113.110299
Stenbæk J, Löf D, Falkman P, Jensen B, Cárdenas M (2017) An alternative anionic bio-sustainable anti-fungal agent: investigation of its
mode of action on the fungal cell membrane. J Colloid Interface
Sci 497:242–248. https://doi.org/10.1016/j.jcis.2017.03.018
Szkolnik M, Gilpatrick JD (1969) Apparent resistance of Venturia
inaequalis to dodine in New York apple orchards. Plant Dis
Report 53:861–864
Tavano L, Pinazo A, Abo-Riya M, Infante MRR, Manresa MAA,
Muzzalupo R, Pérez L (2014) Cationic vesicles based on biocompatible diacyl glycerol-arginine surfactants: physicochemical properties, antimicrobial activity, encapsulation efficiency and drug release. Colloids Surf B Biointerfaces 120:160–167. https://doi.org/
10.1016/j.colsurfb.2014.04.009
Tedersoo L, Sánchez-Ramírez S, Kõljalg U, Bahram M, Döring M,
Schigel D, May T, Ryberg M, Abarenkov K (2018) High-level classification of the fungi and a tool for evolutionary ecological analyses. Fungal Divers 90:135–159. https://doi.org/10.1007/s13225018-0401-0
Terjung N, Loeffler M, Gibis M, Salminen H, Hinrichs J, Weiss J (2014)
Impact of lauric arginate application form on its antimicrobial activity in meat emulsions. Food Biophys 9:88–98. https://doi.org/10.
1007/s11483-013-9321-4
Tezel U, Pavlostathis SG (2015) Quaternary ammonium disinfectants:
microbial adaptation, degradation and ecology. Curr Opin
Biotechnol 33:296–304. https://doi.org/10.1016/j.copbio.2015.03.
018
Tóth A, Petróczy M, Hegedűs M, Nagy G, Lovász C, Ágoston J,
Palkovics L (2012) Development of plant protection technology
against sour cherry anthracnose. In: György J, Kövics GJ, Dávid I
(eds) 6th International Plant Protection Symposium at University of
Debrecen. Journal of agricultural sciences. Acta agraria
debreceniensis, Debrecen, Hungary, pp 54–59
Tripathy DB, Mishra A, Clark J, Farmer T (2018) Synthesis, chemistry,
physicochemical properties and industrial applications of amino acid surfactants: a review. Comptes Rendus Chim 21:112–130. https://
doi.org/10.1016/j.crci.2017.11.005
Tsui C, Kong EF, Jabra-Rizk MA (2016) Pathogenesis of Candida
albicans biofilm. Pathog Dis 74:ftw018. https://doi.org/10.1093/
femspd/ftw018
Tyagi S, Tyagi VK (2014) Novel cationic gemini surfactants and methods
for determination of their antimicrobial activity—review. Tenside
Surfactants Deterg 51:379–386. https://doi.org/10.3139/113.110319
van der Rest ME, Kamminga AH, Nakano A, Anraku Y, Poolman B,
Konings WN (1995) The plasma membrane of Saccharomyces
cerevisiae: structure, function, and biogenesis. Microbiol Rev 59:
304–322
Vieira DB, Carmona-Ribeiro AM (2006) Cationic lipids and surfactants
as antifungal agents: mode of action. J Antimicrob Chemother 58:
760–767. https://doi.org/10.1093/jac/dkl312
Wang L-F, He D-Q, Tong Z-H, Li W-W, Yu H-Q (2014) Characterization
of dewatering process of activated sludge assisted by cationic surfactants. Biochem Eng J 91:174–178. https://doi.org/10.1016/j.bej.
2014.08.008
Weber DJ, Rutala WA (2013) Self-disinfecting surfaces: review of current
methodologies and future prospects. Am J Infect Control 41:S31–
S35. https://doi.org/10.1016/j.ajic.2012.12.005
Yagura Y, Kirinuki T, Matsunaka S (1984) Mode of action of the fungicide guazatine in Alternaria kikuchiana. J Pestic Sci 9:425–431.
https://doi.org/10.1584/jpestics.9.425
Yoder KS, Klos EJ (1976) Tolerance to dodine in Venturia inaequalis.
Phytopathology 66:918–923