Galleria Mellonella As An Infection Model: An In-Depth Look at Why It Works and Practical Considerations For Successful Application

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Pathogens and Disease, 78, 2020, ftaa056

doi: 10.1093/femspd/ftaa056
Advance Access Publication Date: 0 2020
Minireview

MINIREVIEW

Galleria mellonella as an infection model: an in-depth


look at why it works and practical considerations for
successful application
Monalessa Fábia Pereira1, *,† , Ciro César Rossi2,† , Giarlã Cunha da Silva3 ,
Jéssica Nogueira Rosa3 and Denise Mara Soares Bazzolli3, *,‡
1
Laboratório de Bioquı́mica e Microbiologia, Departamento de Ciências Biológicas, Universidade do Estado de
Minas Gerais, 36800-000, Carangola, MG, Brazil, 2 Laboratório de Microbiologia Molecular, Departamento de
Microbiologia Médica, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro,
21941-901, Rio de Janeiro, RJ, Brazil and 3 Laboratório de Genética Molecular de Bactérias, Instituto de
Biotecnologia Aplicada à Agropecuária—BIOAGRO, Departamento de Microbiologia, Universidade Federal de
Viçosa, 36570-900, Viçosa, MG, Brazil

Corresponding authors: Denise Mara Soares Bazzolli, Tel: +55-31-3612-2454; E-mail: dbazzolli@ufv.br, and Monalessa Fábia Pereira; E-mail:
monalessa.pereira@uemg.br

One sentence summary: In this review, we describe, with examples, the advantages that make Galleria mellonella a versatile infection model to study
host–bacterial pathogen interactions, and discuss practices aimed at successful experiments.

These authors contributed equally to the work.
Editor: Juliana Campos Junqueira

Denise Mara Soares Bazzolli, http://orcid.org/0000-0002-0371-6966

ABSTRACT
The larva of the greater wax moth Galleria mellonella is an increasingly popular model for assessing the virulence of bacterial
pathogens and the effectiveness of antimicrobial agents. In this review, we discuss details of the components of the G.
mellonella larval immune system that underpin its use as an alternative infection model, and provide an updated overview
of the state of the art of research with G. mellonella infection models to study bacterial virulence, and in the evaluation of
antimicrobial efficacy. Emphasis is given to virulence studies with relevant human and veterinary pathogens, especially
Escherichia coli and bacteria of the ESKAPE group. In addition, we make practical recommendations for larval rearing and
testing, and overcoming potential limitations of the use of the model, which facilitate intra- and interlaboratory
reproducibility.

Keywords: Galleria mellonella; animal alternative model; microbial pathogenicity; antimicrobial therapy; 3Rs

INTRODUCTION 2016; Cutuli et al. 2019). Over 2000 scientific articles have been
published in PubMed on G. mellonella so far, of which 271 were
The larva of the greater wax moth Galleria mellonella is well
published in 2019 alone, demonstrating the growing popular-
accepted by the scientific community worldwide (Fig. 1) as an
ity of this infection model. In particular, there has been an
experimental model to study host–pathogen interactions and
expansion in the diversity of bacterial pathogens studied, and G.
the effectiveness of antimicrobial agents (Tsai, Loh and Proft
mellonella–pathogen infection models have made an invaluable

Received: 25 June 2020; Accepted: 18 September 2020



C FEMS 2020. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com

1
2 Pathogens and Disease, 2020, Vol. 00, No. 00

Figure 1. Locations and representativity (%) of institutions that conduct studies with G. mellonella. To build this worldwide overview of G. mellonella research, we
considered the country of the institution to which the corresponding author belongs. The analysis was made after obtaining data from a bibliographic search carried
out in July 2020 at PubMed (MEDLINE database) with articles containing the keyword ‘Galleria mellonella’.

contribution to research with human and veterinary bacterial virulence of microbial pathogens and produce results compara-
pathogens (Tsai, Loh and Proft 2016; Cools et al. 2019). ble to those obtained with mammalian systems (Sheehan et al.
The use of the G. mellonella model reinforces the impor- 2018).
tance of applying the ‘3Rs’ principles (replacement, reduction Although results depend highly on the target specificity of
and refinement) in animal experimentation, aiming to substi- the microorganisms’ arsenal of virulence, G. mellonella’s rel-
tute vertebrate infection models and, consequently, reduce the atively advanced system of antimicrobial defenses makes it
number of mammals used in research. As an invertebrate, G. mel- a suitable and informative infection model (López Hernández
lonella is not included in animal welfare legislation and ethics et al. 2015). Studies have shown that some virulent bacterial
guidelines (Graham and Prescott 2015; Hubrecht and Carter strains are lethal for G. mellonella as they are for mammalian
2019). hosts, especially mice (Seed and Dennis 2008; Pereira et al. 2015;
The advantages of using the G. mellonella model are numer- Propst et al. 2016). Moreover, for some important pathogenic
ous (Fig. 2). Rearing G. mellonella is less expensive and easier to species, histopathological effects and cellular responses found
maintain than mammalian models, and does not require major in G. mellonella resemble those found in human infections.
adaptations to the laboratory’s infrastructure. The large size of For example, Staphylococcus aureus infection results in nod-
the larvae (12–20 mm) ensures easy handling, and facilitates the ule formation in G. mellonella with similar structures to those
collection of tissues and analysis. The large size of the larvae found in human abscesses (Sheehan, Dixon and Kavanagh
also allows the precise quantification of the inocula, which can 2019). Likewise, histopathological effects caused by Mycobac-
be injected directly into the larval hemocoel (Ramarao, Nielsen- terium abscessus include granulomatous structures as seen in
Leroux and Lereclus 2012; Cook and McArthur 2013). Galleria mel- humans infected with this bacterium (Meir et al. 2018). In addi-
lonella’s short life cycle makes it suitable for high-throughput tion to similar pathological effects being found in G. mellonella to
studies (Tsai, Loh and Proft 2016). From our decade-long expe- those in conventional mammalian infection models, increased
rience with this insect, a good-quality diet and careful breeding levels of survival in response to antibacterial treatment have
can lead to a life cycle of 40–60 days. also been observed in G. mellonella and mice (Jeon et al. 2019;
Some physiological and immunological characteristics of G. Leshkasheli et al. 2019).
mellonella are fundamental for the success of the model. Lar- Despite the numerous advantages offered by G. mellonella for
vae can be kept at 37◦ C, equivalent to the body temperature of the study of Gram-positive and Gram-negative bacteria, it is
mammalian hosts. As temperature has been shown to affect the important to be aware of experimental variables that can influ-
expression of microbial virulence factors, this feature is impor- ence the outcome of the study. In addition, the limitations of the
tant when assessing virulence of mammal pathogens (Tsai, Loh model must be recognized, so that the results can be properly
and Proft 2016; Twittenhoff et al. 2020). Despite not having an interpreted. This review will address the advantages and appli-
adaptive immune response, the immune system of G. mellonella cations of the model to study bacterial pathogens, and discuss
shares many similarities with the mammalian innate immune issues that we consider to be of great relevance for strengthen-
response and, as a consequence, can be exploited to assess the ing the use of G. mellonella as an alternative infection model.
Pereira et al. 3

Figure 2. Advantages offered by the G. mellonella alternative infection model to study bacterial pathogens. Several aspects of the G. mellonella larva support its application
in in vivo experiments: a high number of repetitions will increase the statistical support of the study; the fact that the larva is relatively large (12–20 mm) allows easy
handling and extraction of tissues and fluids, as well as inoculation of precisely quantified substances and microorganisms directly in the hemocoel; the larvae can be
reared in the laboratory in small containers, provided with natural air flow, without the need for major adaptations in the laboratory structure; since the larval innate
immune system shares similarities with that of mammals, experiments with the insect can, to some extent, replace experiments with vertebrates; a good-quality diet
and careful breeding can lead to a shorter life cycle of 40–60 days; and the larvae can be kept at 37◦ C, equal to the body temperature of mammals and suitable to test
several bacterial pathogens. These advantages allow faster screening of varied compounds with antimicrobial activity before testing in vertebrates.

THE IMMUNE SYSTEM OF G. MELLONELLA They play a major role in the insect’s innate immunity, given
AND THE SUCCESS OF THE MODEL their broad-spectrum microbicidal activity. So far, 18 known or
putative AMPs have been identified, some of which are similar
A primary reason for the success of the G. mellonella model to molecules characterized in mammals, namely: two types of
in the study of microbial virulence is that its immune system cecropins, a heliocin-like peptide, an inducible serine protease
shares a high degree of structural and functional similarity with inhibitor 2, galiomycin, gallerimycin, gloverin, Gm anionic pep-
that of the innate immune system of vertebrates. The immune tides 1 and 2, Gm proline-rich peptides 1 and 2, lysozyme, five
response of G. mellonella consists of two tightly interconnected types of moricin-like peptides and X-tox (Brown et al. 2009; Tsai,
components: cellular (cell-mediated) and humoral responses Loh and Proft 2016; Sheehan et al. 2020).
(Browne, Heelan and Kavanagh 2013). Galleria mellonella produces several opsonins that recognize
The cellular response is mediated by hemocytes, cells analo- and bind to conserved microbial components such as the
gous to human phagocytes, involved with phagocytosis, encap- lipopolysaccharides (LPS), lipoteichoic acid (LTA) and peptidogly-
sulation and nodulation of the invading agent (Fig. 3). To date, six can, similar to the pattern recognition receptors (PRRa) found in
types of hemocytes have been identified in G. mellonella: prohe- mammals.
mocytes, plasmatocytes, granular cells, coagulocytes, spherulo- The opsonin Apolipophorin-III (ApoLp-III) has a high affin-
cytes and oenocytoids. Plasmatocytes and granular cells are the ity for hydrophobic ligands, such as LPS and LTA. ApoLp-III has
most abundant hemocytes in the hemolymph (Wu et al. 2016). a multifunctional role in the immune system of G. mellonella,
Hemocytes are found free in the hemolymph or attached to being able to stimulate the production of superoxide by hemo-
internal organs, such as the digestive tract, fat body and heart cytes, increase the production of cecropin and act synergis-
surface of the insect. The concentration of hemocytes in the tically with lysozyme, thus increasing its antimicrobial activ-
hemolymph varies during the life of the insect and in response ity (Maravilla et al. 2020). ApoLp-III is also involved in recog-
to pathogens (Browne, Heelan and Kavanagh 2013; Pereira et al. nizing/differentiating the type of pathogen (e.g. Gram-positive
2015; Arteaga Blanco et al. 2017). and Gram-negative bacteria, yeasts and filamentous fungi), and
The humoral response involves soluble effector molecules assembling an adequate immune response (Stączek et al. 2018).
such as antimicrobial peptides (AMPs), complement-like pro- The peptidoglycan recognition protein (PGRP), another
teins (opsonins), melanin and products of proteolytic cascades, opsonin, recognizes peptidoglycan (Wang et al. 2019). In some
which immobilize or kill pathogens (Eleftherianos and Revenis insects, PGRPs have been shown to hydrolyze peptidoglycan, but
2011; Sheehan et al. 2018). this has not yet been demonstrated in G. mellonella (Tsai, Loh and
Proteomic studies have shed light on the great diversity and Proft 2016).
dynamics of AMP production (Sheehan, Dixon and Kavanagh GmCP8 is an opsonin produced in the fat body, the midgut
2019; Sheehan et al. 2020), which has been recently shown to be and the integument of G. mellonella. It is secreted in the
regulated, in part, by non-coding microRNAs (miRNAs), as it hap- hemolymph and binds strongly to LPS, LTA and β-1,3-glucan
pens in vertebrates (Mukherjee et al. 2020). AMPs are produced (Kim et al. 2010; Barbaro et al. 2019).
mainly in the fat body, hemocytes, salivary glands and repro- The opsonin hemolin, a member of the immunoglobulin
ductive tract of G. mellonella in response to microbial invasion. superfamily, binds LPS and LTA, and associates with hemocytes
4 Pathogens and Disease, 2020, Vol. 00, No. 00

Figure 3. Major components of G. mellonella innate immune system. Galleria mellonella immune system comprises components of a cellular response and a humoral
response. The cellular response comprehends six types of cells (top left): the prohemocytes, plasmatocytes, granulocytes, spherulocytes, oenocytoids and coagulo-
cytes. Some of these cells (but not only those depicted in the top right of the figure) are involved with clearance of invading agents by phagocytosis, nodulation and
encapsulation. Some cells capable of recognizing molecular patterns associated to pathogens (such as the peptidoglycan and lipopolysaccharides, bottom right) trigger
the phenoloxidase pathway, to produce melanin. Melanin is one of the components of the humoral response of the innate immune system of G. mellonella, which also
includes antimicrobial peptides and opsonins (bottom left).

(Aathmanathan et al. 2018). Hemolin can be expressed in the silk previous results showed how G. mellonella’s cellular and humoral
gland and fat body of G. mellonella, and is upregulated during immune responses are integrated: the site with the strongest
bacterial and fungal infections (Shaik and Sehnal 2009; Shee- melanization (the heart) is also the region with the highest con-
han et al. 2020). Hemolin transcripts are induced by bacteria or centration of hemocytes (Pereira et al. 2015).
purified bacterial components, such as the LPS and phorbol 12- Although innate immune responses are non-specific, they
myristate 13-acetate (Aathmanathan et al. 2018). are the first line of defense against invading pathogens, being
The melanization process is essential for the defense of G. widely distributed throughout the body to maintain homeosta-
mellonella against microbial pathogens, and results in the syn- sis and prevent infections (Sheehan et al. 2018). The lack of
thesis and deposition of melanin around the microbe in the an adaptive immunity in G. mellonella and other insects can be
hemolymph (Kavanagh and Reeves 2004). Melanin production is seen as an advantage for research purposes, since the model
catalyzed by the enzyme phenoloxidase (PO), which is produced allows the study of host–pathogen interactions and related
as the inactive zymogen pro-phenoloxidase (pro-PO) in hemo- innate immunity mechanisms without the interference of adap-
cytes. Therefore, pro-PO is involved in both cellular and humoral tive responses (Kavanagh and Sheehan 2018). Therefore, the
defense. Melanization begins after recognition and phagocytosis G. mellonella model can substitute for more complex vertebrate
of the pathogen, when soluble PRRs coupled to target molecules models, depending on the objective of the study, and when the
on bacterial surfaces trigger the serine protease cascade that absence of the adaptive immune system limits the possibility of
results in the cleavage of pro-PO to PO. The PO catalyzes the oxi- replacing vertebrate models, G. mellonella can be an intermediate
dation of phenols to quinones, which spontaneously polymerize model between in vitro and vertebrate in vivo studies.
to form melanin around invading pathogens (Eleftherianos and
Revenis 2011). As overproduction of PO can lead to the produc-
tion of cell-damaging reactive oxygen species, PO activation is USING G. MELLONELLA TO INVESTIGATE THE
highly controlled by protease inhibitors (Lu et al. 2014). VIRULENCE OF BACTERIAL PATHOGENS
The form and speed with which melanization occurs vary
The potential of a bacterium to cause disease is related to its
according to the virulence of the pathogen and inoculum size
repertoire of virulence factors. In general, virulence factors are
(Pereira et al. 2015; Guerrieri et al. 2019). Typically, melanization
divided into those that: (i) allow entry and establishment of
starts with distinct black spots on the surface of the larval cuti-
infection in the host; (ii) contribute to the evasion of the host’s
cle and, as the infection progresses, the larva can become com-
immune response; and (iii) allow the bacteria to multiply and
pletely melanized. This event correlates with subsequent larval
spread throughout the host or to a new host. However, bacterial
death (Tsai, Loh and Proft 2016). After infection, a more pro-
virulence is complex and multifaceted. Species and strains have
nounced melanization is observed in the dorsal region of the
different sets of virulence genes, and these can be subjected to
larva, comprising the heart (Pereira et al. 2015). This region was
regulation, which makes virulence even more diverse (Diard and
identified as the larva’s new immune tissue, because it is flanked
Hardt 2017; Weigel and Dersch 2018).
by sessile hemocytes that sequester the pathogens in flux in
In this context, G. mellonella has been extensively used to
the hemolymph, and recruit circulating hemocytes to the heart
study bacterial virulence (Fig. 4), which can be evaluated in sev-
region, where they bind to the cardiac muscle and continue to
eral ways after inoculation with a pathogenic microorganism,
phagocytose pathogens (King and Hillyer 2012; Hillyer 2016). Our
such as observing melanization and larval death over time (Kay
Pereira et al. 5

Figure 4. Use of G. mellonella model to study bacterial virulence. The graphics show the diversity and representativeness of Gram-negative and Gram-positive species
that have had their virulence investigated by the G. mellonella model. The data were obtained after a bibliographic search conducted in July 2020 at PubMed (MEDLINE
database) with the keyword ‘Galleria mellonella’, and subsequent individual assessment of the nature of each scientific article.

et al. 2019), density and morphology of hemocytes (Pereira et al. biofilms, by expressing the intercellular polysaccharide adhesin
2015; Sheehan et al. 2019), expression of enzymes and AMPs molecule (PIA) encoded by the ica operon (Haddad et al. 2018; Ma
(Moghaddam et al. 2016) and histopathological effects (Meir et al. et al. 2019).
2018). Some staphylococcal toxins weaken the host’s response, as
Below, we discuss established virulence factors of represen- they degrade host cells, and manipulate both innate and adap-
tative major human and veterinary bacterial pathogens, and tive immune responses, and contribute to S. aureus proliferation
relevant virulent studies in G. mellonella infection models. The (Tam and Torres 2019). Among the main toxins of S. aureus are
information in the text is also summarized and supplemented pore-forming toxins, such as hemolysin, and leukotoxins, e.g.
with the findings for other relevant and well-studied pathogens the Panton–Valentine - PVL (Oliveira, Borges and Simões 2018;
within the ESKAPE group (comprising Enterococcus faecium, Stap Tam and Torres 2019). Staphylococcus aureus also produces a vari-
hylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, ety of virulence factors with enzymatic properties. They can
Pseudomonas aeruginosa, Enterobacter spp.), in Table 1. break down host barriers or molecules for nutrient acquisition,
bacterial survival and dissemination (e.g. coagulase, staphylok-
inase, nucleases, proteases, metalloprotease, serine proteases,
Galleria mellonella studies on the virulence of S. aureus cysteine proteases, hyaluronidase and lipases) (Tam and Torres
2019).
Staphylococcus aureus is the most widely studied Gram-positive
Staphylococcus aureus has a highly clonal population struc-
pathogen in G. mellonella infection models (Fig. 4). Given its
ture with clonal complexes (CCs) comprising closely related,
prevalence and the increasing level of strains resistant to multi-
although not identical, genetic backgrounds (Dayan et al. 2016;
ple drugs, S. aureus is classified by the World Health Organization
Turner et al. 2019). Pérez-Montarelo et al. (2017) analyzed the vir-
(2017) as one of the priority microorganisms in studies for new
ulence of S. aureus strains from five representative CCs associ-
antimicrobial therapies. First, we will discuss some major viru-
ated with endovascular complications, using the G. mellonella
lence factors of S. aureus, and then some studies in G. mellonella
model. This was the first study comparing the virulence of dif-
investigating S. aureus pathogenicity.
ferent S. aureus CCs in vivo, and revealed significant differences
Staphylococcus aureus has a broad repertoire of virulence
between them. CC30 strains exhibited low virulence against
genes, enabling the bacterium to cause several types of diseases,
G. mellonella, corroborating previous studies that had reported
including toxic shock syndrome, sepsis, endocarditis, pneumo-
significant virulence attenuation of CC30 in G. mellonella and
nia, food poisoning, and skin and soft tissue infections (Lee et al.
murine sepsis models (Sharma-Kuinkel et al. 2017), while CC15
2018). Among the most studied virulence factors of this bac-
strains displayed high virulence. Genetic analysis of CC15 strains
terium are surface-associated and secreted proteins (Laabei et al.
revealed that they had specific virulence gene variants, which
2015; Tam and Torres 2019). The collectively called ‘microbial
were likely associated with the phenotype of higher virulence
surface components recognizing adhesive matrix molecules’
against G. mellonella. These CC15 virulence variants were more
(MSCRAMMs) are proteins that mediate the adherence of S.
notable in genes encoding for leukocidins and proteases. One
aureus to host tissues, including clumping factors A and B (ClfA
CC15 strain presented a particularly high virulence phenotype,
and ClfB), fibronectin-binding proteins (FnBPs), collagen-binding
which was correlated with the presence of the lukFS genes, cod-
adhesin (Cna) and elastin-binding protein (Ebps). Staphylococ-
ing for PVL.
cus aureus also maintains its cellular aggregation, including in
6 Pathogens and Disease, 2020, Vol. 00, No. 00

Table 1. Overview of some studies to assess several aspects of bacterial virulence and treatment with antimicrobial compounds, with emphasis
on species of the ESKAPE group and Escherichia coli.

Bacterial species Type of study Reference

Acinetobacter Comparison of virulence of different strains (Chusri et al. 2019)


baumannii
Study of mutant strains for important virulence factors (Weidensdorfer et al. 2019;
Niu et al. 2020)
Discovery of a new hypervirulent strain (Zhou et al. 2020)
Combination of drugs with adjuvants to treat infections caused by MDR strains to (Minrovic et al. 2018)
reduce colistin dose
Antimicrobial peptide therapies for the treatment of infections caused by MDR (Chung et al. 2016)
strains
Plant natural compounds to control bacterial infections caused by MDR strains (Betts et al. 2017)
Enterobacter sp. Comparison of virulence of different subpopulations of clinical isolates (Brust et al. 2019)
Characterization of virulence of a representative strain of a new species of the (Pati et al. 2018)
genus Enterobacter
Combination of therapies to treat infections caused by MDR strains (Betts et al. 2014; Yang et al.
2016)
Control of infections with bacteriophage therapy (Manohar, Nachimuthu and
Lopes 2018)
Enterococcus sp. Comparison of virulence among vancomycin-resistant clinical strains (Lam et al. 2013)
Study of mutant strains for important virulence factors (Salze et al. 2020; Zheng et al.
2020)
Antimicrobial photodynamic therapy for the treatment of infections caused by (Chibebe et al. 2013)
vancomycin-resistant strains
Combination of antimicrobial therapies to treat infections (Thieme et al. 2020)
Escherichia coli Understanding of regulatory networks that modulate the expression of LEE (Morgan, Ortiz and Riordan
virulence factors in EHEC with studies of gene expression and mutant virulence 2014; De la Cruz et al. 2016)
Comparison of virulence among clinical strains of typical and atypical EAEC (Guerrieri et al. 2019)
Assessment of regulation mechanisms of innate immunity against infections (Mukherjee et al. 2020)
caused by UPEC strains by miRNAs
Combination of antimicrobial therapies to treat infections caused by MDR and (Brochado et al. 2018)
XDR strains
Antimicrobial peptide therapies for the treatment of infections caused by MDR (Vergis et al. 2019)
strains
Control of infections with bacteriophage therapy (Manohar, Nachimuthu and
Lopes 2018)
Klebsiella pneumoniae Study of mutant strains for important virulence factors (Insua et al. 2013; Mills et al.
2017)
Discrimination of strains regarded as highly virulent from less virulent strains (Insua et al. 2013)
Comparison of virulence of colistin-resistant isolates (Esposito et al. 2018)
Discrimination of virulence level of hvKP strains (Shi et al. 2018; Li et al. 2020)
Combination of therapies to treat infections caused by KPC strains (Nath et al. 2018)
Control of infections with bacteriophage therapy (Manohar, Nachimuthu and
Lopes 2018; Thiry et al. 2019)
Staphylococcus Comparison of virulence of different clonal complexes (CCs) of clinical relevance (Pérez-Montarelo et al. 2017)
aureus
Assessment of the relationship between the extracellular proteome and the (Zhao et al. 2019)
virulence of strains
Characterization of infection development (Sheehan, Dixon and
Kavanagh 2019)
Combination of antimicrobial therapies to treat infections (Li et al. 2019)
Antimicrobial peptide therapies to treat infections caused by MRSA strains (Yuan et al. 2019)
Use of plant natural compounds to control bacterial infections caused by MDR (Knidel et al. 2019; Bezerra
strains Filho et al. 2020)
Control of infections with bacteriophage therapy (Tkhilaishvili et al. 2020)
Pseudomonas Comparison of virulence of different strains (Medina-Rojas et al. 2020)
aeruginosa
Study of mutant strains for important virulence factors (Klein et al. 2019; Lo Sciuto
et al. 2019)
Screening for virulence of new strains (Kaszab et al. 2019)
Combination of drugs with adjuvants to treat infections caused by MDR and XDR (Brochado et al. 2018; Idowu
strains et al. 2019)
Antimicrobial peptide therapies for the treatment of infections caused by MDR (Zheng et al. 2017)
strains
Control of infections with bacteriophage therapy (Forti et al. 2018)
Actinobacillus Discriminate the virulence of different isolates (Pereira et al. 2018)
pleuropneumoniae
Study of mutant strains for a pleiotropic regulator (Pereira et al. 2015)
Pereira et al. 7

Another study, carried out by Graf et al. (2019), evaluated in agent of swine pleuropneumonia, a contagious disease associ-
G. mellonella the activity of virulence factors from the extracel- ated with high morbidity and mortality in swine herds.
lular matrix of biofilms of S. aureus, an essential determinant of
the colonization of medical devices that make this bacterium
Galleria mellonella studies on the virulence of E. coli
one of the major causes of nosocomial infections. These fac-
tors were identified by a multiomic approach and included cap- The diversity of E. coli is well documented in terms of its genet-
sule proteins, hemolysins, leukotoxins and lipases. The cell-free ics and ability to live as a commensal or pathogen in different
biofilm supernatant containing these factors was injected into animal and human hosts. This wide diversity arises, in part,
the hemolymph of G. mellonella larvae, leading to a high mortal- because of constant changes that occur in the E. coli genome,
ity rate and demonstrating the utility of the model in the study due to a continuous flow of genetic insertions, deletions and
of virulence factors secreted by S. aureus. horizontal gene transfer. While the E. coli genome encodes
The extracellular proteome (exoproteome) of S. aureus is a ∼4000–6500 proteins, the core genome of the species comprises
main reservoir of virulence factors, showing substantial hetero- only 2000 genes. The additional genes determine commensal-
geneity for different clonal lineages (Ziebandt et al. 2010; Zhao ism, species specificity, pathotypes and antibiotic susceptibility
et al. 2019). A study by Zhao et al. (2019) showed that the vir- (Croxen and Finlay 2010; Poolman 2017).
ulence of S. aureus spa-type t437 may be directly related to its Pathogenic E. coli strains can be divided in two groups: (i)
exoproteome profile, and that the G. mellonella killing assay can strains that cause diarrheal intestinal disease (Enteropathogenic
accurately differentiate between these profiles. Certain proteins E. coli [EPEC], Enterotoxigenic E. coli [ETEC], Enterohemorrhagic E.
such as IsaA, IsdB, IsdA, IsdE, IsdH and Chitinase B were related coli [EHEC], Enteroaggregative E. coli [EAEC], Enteroinvasive E. coli
to the killing of larvae. The IsaA protein, a major antigen of S. [EIEC] and Diffusely Adherent E. coli [DAEC]), and (ii) strains that
aureus, was exclusive to the exoproteome of groups that were usually cause disease outside the intestinal tract (Extraintesti-
highly virulent against G. mellonella, in line with the attenuated nal Pathogenic E. coli [ExPEC]), being Uropathogenic E. coli (UPEC)
activity of an isaA deletion mutant. Chitinase B was also exclu- and Neonatal Meningitis E. coli (NMEC), the most common ExPEC
sive to the exoproteome of the highly virulent groups, suggesting (Croxen and Finlay 2010; Poolman 2017).
that this enzyme may impact immune evasion and infection in The pathogenic strains of E. coli have a wide arsenal of vir-
G. mellonella, similar to its activity in human hosts (da Silva et al. ulence strategies; many are pathotype specific, but some are
2009). Mutants for isdA or isdB were not attenuated in G. mel- required for virtually all of them. Adhesion to host cells, fre-
lonella, showing that these siderophores may not be relevant to quently achieved by fimbriae or pili, is required by all patho-
cause infection in this insect. types, except EIEC. After adhesion, E. coli subverts host cell pro-
The use of G. mellonella for studying S. aureus is endorsed by cesses, often using secreted proteins and manipulates host cell
the strong interaction of this pathogen with the humoral and signaling pathways, resulting in coordinated host cell invasion,
cellular immune response of this insect. Staphylococcus aureus evasion of host immune responses, efficient colonization and
induces the expression of a variety of peptides related to the disease (Croxen and Finlay 2010; Khairy et al. 2019). Seeking to
immune system and also forms nodules, which are similar in investigate E. coli virulence, G. mellonella has been widely used
structure and function to abscesses commonly found during for studying commensal strains (Mukherjee et al. 2020), as well
S. aureus skin and soft tissue infections in humans (Sheehan, as those related to diarrheal intestinal disease (De la Cruz et al.
Dixon and Kavanagh 2019). This shows that S. aureus virulence 2016; Guerrieri et al. 2019), and those responsible for extraintesti-
factors are recognized by the larval immune system, and its nal infections (Williamson et al. 2014; Mukherjee et al. 2020).
elimination occurs through mechanisms similar to those of its Human infections with EHEC can induce hemorrhagic coli-
natural (human) host. tis (bloody diarrhea) and the life-threatening hemolytic uremic
It is noteworthy that the most common, accurate and reliable syndrome, as a result of the production of Shiga toxin (Stx), the
way to assess the virulence of bacteria in G. mellonella is through main virulence factor of EHEC. There are two subgroups of Stx
the inoculation of the microorganism directly in the insect’s (Stx1 and Stx2), found in various combinations (Poolman 2017).
hemolymph. This practice, however, saves the pathogen from EHEC pathogenicity is also involved with its ability to colonize
obstacles that are usually present in the initial stages of infec- the gut mucosa, leading to the development of intestinal attach-
tion, that are overcome by factors involved in adherence, break- ing and effacing (AE) lesions (Nataro and Kaper 1998). Most pro-
ing barrier integrity and colonization. Since these obstacles are teins required for the formation of AE lesions are encoded in a
absent in G. mellonella assays, the bacteria are readily confronted pathogenicity island called locus of enterocyte effacement (LEE),
by the larval innate immune system’s components. Therefore, which encodes a type III secretion system (T3SS) and numerous
studies on virulence factors involved in the early stages of infec- effector proteins (De la Cruz et al. 2016; Schwidder, Heinisch and
tion, as well as those factors that have very specific targets, need Schmidt 2019).
special attention during experimentation, so the results can be Studies on gene expression and virulence of mutants against
properly interpreted. G. mellonella have helped understanding regulatory networks
Most G. mellonella studies with bacterial pathogens focus that modulate the expression of LEE virulence factors. De la Cruz
on Gram-negative bacteria (Fig. 4). Next, we discuss the recent et al. (2016) showed that the CpxA sensor kinase, which con-
advances in understanding the virulence of two of these trols E. coli envelope production, is related to the complex reg-
microorganisms: E. coli and K. pneumoniae. Both are Enterobac- ulatory network of the LEE. This was concluded after the EHEC
teriaceae with high levels of resistance, among the most impor- cpxA mutant showed reduced expression of LEE genes and was
tant opportunistic human pathogens and are also classified by significantly deficient in killing G. mellonella. The role of TolA in
the World Health Organization (2017) as top priorities in stud- EHEC pathogenesis and regulation of virulence genes contained
ies aiming at new antimicrobial therapies. Finally, we present in LEE (i.e. T3SS, common pilus [ecpA] and motility [fliC]) was ver-
the main findings obtained by our group when studying the vet- ified by combining studies of gene expression and investigation
erinary pathogen Actinobacillus pleuropneumoniae, the causative of tolA mutants, which were substantially attenuated in G. mel-
lonella (Morgan, Ortiz and Riordan 2014).
8 Pathogens and Disease, 2020, Vol. 00, No. 00

EAEC is another type of pathogenic E. coli related to diar- three decades in countries of the Asian Pacific Rim (Russo and
rheal intestinal disease and insights into its virulence have been Marr 2019). The hvKP strains are more virulent than cKP, which
obtained with the use of G. mellonella (Jønsson et al. 2017; Guer- makes their identification critical for patient care and epidemi-
rieri et al. 2019). This bacterium is the leading cause of acute ologic studies (Li et al. 2020; Russo and Macdonald 2020).
and persistent diarrhea in all ages worldwide (Okhuysen and Initially, hvKP strains were correlated with the extreme-
DuPont 2010; Spano et al. 2017). Although the pathogenesis of stickiness ‘hypermucoviscous’ phenotype. However, it is now
EAEC is not clear due to the heterogeneity of strains, it is sug- known that not all hvKP strains are hypermucoviscous, and
gested that, after infection, there is an initial adhesion to the some cKP strains have this characteristic, suggesting that hyper-
intestinal mucosa, biofilm formation, induction of inflammatory mucoviscosity and hypervirulence are two different phenotypes
responses and release of toxins (Okeke et al. 2000; Jensen et al. of K. pneumoniae (Catalán-Nájera, Garza-Ramos and Barrios-
2014). The presence of the AggR regulon, a regulator of chro- Camacho 2017). Although early hvKP isolates were sensitive to
mosomal and plasmid-encoded virulence factors, distinguishes antimicrobials, MDR-hvKP strains have been reported, raising
typical from atypical EAEC. It has been suggested that typical concerns about treating infections caused by these bacteria (Shi
EAECs are more virulent than atypical EAECs (Sarantuya et al. et al. 2018; Wyres et al. 2019).
2004; Morin et al. 2013). In vivo tests with G. mellonella were made Genotypic markers, such as peg-344, iroB, iucA, rmpA and
by Guerrieri et al. (2019), indicating that the virulence of EAEC rmpA2, present on the hvKP virulence plasmid can be used to
strains seems to be related, in part, to the AggR regulon, and identify hvKP; however, some low-virulence strains also carry
atypical EAEC strains can be as virulent as typical strains. these markers (Russo and Marr 2019; Li et al. 2020). Several
Studies with G. mellonella have also contributed to a bet- investigations have examined whether the K1 and/or K2 cap-
ter understanding of ExPEC virulence. Although they are more sule types enhanced virulence compared with other types. How-
commonly associated with urinary tract infections, ExPEC ever, although it is possible that the capsule type could modu-
strains cause a range of serious infections, including meningi- late hvKP virulence, hypervirulence is not exclusive to these two
tis, pneumonia and bacteremia. These bacteria have an array types of capsule (Russo and Marr 2019).
of virulence-associated genes (e.g. adhesins, invasins, toxins, The need for an in-depth investigation of K. pneumoniae viru-
siderophores and polysaccharide capsules), often encoded by lence is evident, and G. mellonella has been extensively used for
pathogenicity islands (Poolman 2017; Klein and Hultgren 2020). studying cKP (Insua et al. 2013; Mills et al. 2017; Esposito et al.
Williamson et al. (2014) validated the G. mellonella model for 2018) and hvKP (Shi et al. 2018; Li et al. 2020; Russo and Macdon-
studying ExPEC strains in an analysis that observed that iso- ald 2020).
lates with higher amounts of virulence genes killed larvae sig- Insua et al. (2013) showed that G. mellonella could discrim-
nificantly faster than isolates with lower virulence genes scores. inate strains regarded as highly virulent from others less vir-
In contrast, no relationship was found between the virulence ulent, in addition to pinpoint differences among highly viru-
factor (VF) score and LD50 for ExPEC strains associated with ade- lent strains. They also showed that mutants lacking capsule
nomatous polyps (Ambrosi et al. 2019). Therefore, a high VF score polysaccharides, lipid A decorations, or the outer membrane
does not necessarily correlate with larval mortality. The results proteins OmpA and OmpK36, previously known to be attenuated
suggested that the presence of other genes, either not investi- in the mouse pneumonia model, were also attenuated in G. mel-
gated or unknown, or even regulatory mechanisms, may be nec- lonella. This study also showed that G. mellonella infection with
essary for the complete expression of virulence genes. K. pneumoniae might lead to some of the main characteristics
Studies of G. mellonella infection with UPEC led to the discov- of Klebsiella-induced pneumonia (i.e. cell death associated with
ery of new miRNAs involved with the epigenetic reprograming of bacterial replication, prevention of phagocytosis and attenua-
the larval innate immunity. These miRNAs were not expressed tion of host defense responses, mainly the production of antimi-
when the infection was carried out with commensal strains, crobial factors).
suggesting a novel therapeutic application to treat urinary tract A variable virulence profile was also observed by Esposito
infections in humans (Mukherjee et al. 2020). et al. (2018), in a study with strains of K. pneumoniae resistant to
colistin, a phenotype mainly regulated by modifications in LPS
structure and ionic charge, which is controlled by the PhoQ/PhoP
Galleria mellonella studies on the virulence of K. two-component system. This study observed a variable viru-
lence profile among the isolates, and showed that the MIC of
pneumoniae
colistin was correlated with the values of lethal doses against G.
Klebsiella pneumoniae is another increasingly problematic oppor- mellonella.
tunistic pathogen, mainly due to its high rates of antibiotic resis- Klebsiella pneumoniae strains can present a great variability in
tance, and difficulty in treatment. The emergence and spread virulence and it is difficult to correlate certain genetic charac-
of multidrug-resistant (MDR) K. pneumoniae is a serious public teristics with virulence in vivo (Li et al. 2020). One of the biggest
health threat, which is aggravated by the increased prevalence challenges in studying the virulence of this pathogen is to clearly
of colistin resistance (Pitout, Nordmann and Poirel 2015; Pet- define hvKP strains and their level of virulence (Shi et al. 2018; Li
rosillo, Taglietti and Granata 2019). Although the mechanisms et al. 2020). Many groups have attempted to standardize G. mel-
of K. pneumoniae antimicrobial resistance are widely studied, lonella tests to access hvKP virulence (Li et al. 2020; Russo and
its virulence mechanisms are still poorly understood, with the Macdonald 2020). However, not all strains characterized as hvKP
capsule, fimbriae, LPS and siderophores (i.e. enterobactin, aer- are highly virulent in G. mellonella, and although the presence of
obactin, salmoquelin and yersiniabactin) being the most studied some genes associated with the hypervirulence phenotype do
(Wand et al. 2013; Mills et al. 2017). correlate positively with larval mortality, other virulence factors
Klebsiella pneumoniae strains are divided into classical (cKP) (which may target molecules absent in G. mellonella) do not.
and hypervirulent (hvKP). Currently, most K. pneumoniae infec- Thus, so far, the G. mellonella model alone is not capable of
tions are caused by cKP strains, however, the incidence of infec- accurately differentiating strains of hvKP and cKP, a possible
tions due to hvKP has been steadily increasing over the past
Pereira et al. 9

consequence of the complex genetic background of clinical iso- Combining drugs with adjuvants is another strategy to
lates (Shi et al. 2018; Li et al. 2020). The fact that some cKP strains enhance the effectiveness of antibiotics and delay the emer-
are highly virulent in G. mellonella indicates that the larva is sen- gence of resistance. Idowu et al. (2019) observed, in G. mellonella,
sitive to some virulence factors that are still unknown, and fur- that the non-ribosomal tobramycin–cyclam conjugate confers
ther investigation is needed to discover these factors (Li et al. a potent adjuvant property that restores the complete antibi-
2020). Even so, the model has proved to be an excellent tool for otic activity of meropenem and aztreonam against carbapenem-
the study of mutant strains of K. pneumoniae. The advance of resistant P. aeruginosa, presenting a way to further preserve the
genomic studies and functional genetic analysis will hopefully therapeutic utility of β-lactam antibiotics.
improve our understanding of K. pneumoniae virulence. Colistin, one of the last resources to treat Gram-negative MDR
infections, has recognized high renal and neurological toxicity.
In view of the growing need to use this antibiotic, the use of adju-
Virulence of A. pleuropneumoniae vants in clinical practice could possibly decrease the dosage of
the medication. In this context, Minrovic et al. (2018) presented
Studies performed by our group showed the suitability of the a new urea-containing class of 2-aminoimidazole-based adju-
G. mellonella model to discriminate isolates and different strains vants that potentiates colistin activity against A. baumannii. The
(wild type and mutant) of the pig pathogen A. pleuropneumoniae adjuvants allowed a 1000-fold reduction in the MIC of colistin in
presenting varied levels of virulence (Pereira et al. 2015, 2018). vitro, and showed efficacy in G. mellonella, representing the first
Although the virulence of this pathogen is mostly related to step to validate the potential of these adjuvants to reduce the
the production of pore-forming RTX toxins, we observed that colistin dose.
mutants for the chaperone Hfq are attenuated in the model, as In a much broader study, a pairwise drug combination
seen in pigs, the natural host. Since Hfq is a global regulator of approach was evaluated by Brochado et al. (2018), with dozens
the activity of small regulatory RNAs (sRNAs) in this bacterium, of antibiotics of all major classes, food additives and adjuvants,
our studies indicate that sRNAs regulate virulence in A. pleurop- to assess the antagonism or synergistic activity of almost 3000
neumoniae, though the mechanisms have yet to be determined drug combinations against E. coli, Salmonella enterica serovar
(Pereira et al. 2015; work in progress). Typhimurium and P. aeruginosa. After in vitro experiments, syn-
ergistic combinations were tested in vivo against MDR strains
of these pathogens in the G. mellonella model. In addition to
identifying drug combinations that were effective against these
USING G. MELLONELLA TO EVALUATE THE
bacteria, this study observed that synergism is more common
EFFICACY OF ANTIMICROBIAL THERAPIES
between compounds that target different steps of the same pro-
Treating infections caused by MDR bacteria is a major pub- cess, in a manner that is mostly species specific, revealing a
lic health challenge worldwide. In addition to the increase of great potential for narrow-spectrum therapies.
bacterial resistance to currently available antimicrobials, there AMPs are another promising source for the development of
has been little progress in the development of new antimicro- therapies against antibiotic resistance. These, used alone or in
bials, increasingly limiting therapeutic options (Nathan 2020). combination with antibiotics, have been tested in G. mellonella to
The Gram-negative bacteria E. coli, K. pneumoniae, P. aeruginosa, treat infections by A. baumannii, E. coli, P. areuginosa and S. aureus
A. baumannii and Neisseria gonorrhoeae, and the Gram-positive S. (Chung et al. 2016; Zheng et al. 2017; Vergis et al. 2019; Yuan et al.
aureus and Enterococcus spp. have all been identified as critical 2019).
pathogens with particularly high rates of resistance to antibi- Yuan et al. (2019), for example, evaluated the antimicrobial
otics, resulting in a reducing pool of treatments available for activity of the AMP japonicin-2LF, isolated from frog skin secre-
these organisms (CDC 2019). In this context, in addition to study- tion, against bacterial cystic fibrosis isolates. The AMP increased
ing bacterial virulence, G. mellonella has been extensively used the survival of G. mellonella infected with MRSA and was shown
to evaluate the efficacy of several types of antimicrobial thera- to increase the permeability of the bacterial membrane.
pies (Fig. 5), against Gram-negative and Gram-positive bacteria Zheng et al. (2017) studied the AMP cecropin A2, produced by
(Fig. 6). Aedes aegypti, and its activity against P. aeruginosa. In addition
Aiming at overcoming antimicrobial resistance and decreas- to increasing the survival of G. mellonella larvae infected with
ing the dose of individual drugs, and therapies with combination P. aeruginosa when applied alone, cecropin A2 also had a syner-
of drugs, especially those already used in clinical practice, have gistic effect with tetracycline. This synergism was attributed to
been suggested and evaluated in G. mellonella (Cutuli et al. 2019). the fact that cecropin A2 permeabilizes the bacterial membrane,
Combination therapies are often recommended to treat infec- facilitating the entrance of tetracycline into the cell.
tions caused by K. pneumoniae producing carbapenemase (KPC), Plant natural compounds have also been widely tested in G.
due to limited treatment options. Using G. mellonella, Nath et al. mellonella to control bacterial infections. Betts et al. (2017) tested
(2018) observed that the combination of ceftazidime/avibactam the efficiency of the natural polyphenols theaflavin and epicat-
with carbapenem should be considered in the treatment of seri- echin, alone and in combination, against six clinical isolates
ous infections caused by KPC. of MDR A. baumannii. The polyphenol combination theaflavin–
Likewise, studies by Li et al. (2019) indicate that the combina- epicatechin produced effective antibacterial activity and showed
tion of linezolid and fosfomycin has synergistic effects against great potential for the treatment of infections caused by MDR A.
S. aureus in G. mellonella, and suggest that high doses of line- baumannii.
zolid and fosfomycin may not be necessary. Thieme et al. (2020) An essential oil isolated from Eugenia brejoensis, a plant from
observed, in G. mellonella, the synergism between ampicillin and the Brazilian semiarid ecosystem, has antimicrobial activity.
ceftriaxone, efficient to treat high E. faecalis inocula. The study Bezerra Filho et al. (2020) showed that, in addition to killing sev-
also questions the benefits of the currently used combination of eral MDR S. aureus strains, subinhibitory concentrations of this
ampicillin and the nephrotoxic antibiotic gentamicin, instead of oil reduced bacterial hemolytic activity, and increased its sus-
the safer ceftriaxone. ceptibility to hydrogen peroxide. When applied to G. mellonella
10 Pathogens and Disease, 2020, Vol. 00, No. 00

Figure 5. Applications of the G. mellonella model to study bacterial pathogens and antimicrobial agents. Depending on the nature of the arsenal of virulence factors of
a given bacterial population, different strains can lead to different patterns of larval melanization and killing (distinct strains are depicted in the top left of the figure
as non-encapsulated cells, encapsulated cells and flagellated cells, respectively); the same applies to the study of mutants for virulence genes (top center). In the face
of the ongoing increase of antimicrobial resistance, G. mellonella has been extensively used to evaluate alternative therapies to combat bacterial infections, including
the study of antimicrobial activity of classical drugs or the combination of drugs (top right); the activity of natural compounds, mostly extracted from plants (bottom
left); the combination of classical drugs with adjuvants (bottom center); and the activity of bacteriophages (phage therapy, bottom right).

Figure 6. Use of G. mellonella model to evaluate antimicrobial therapies. The graphics show the diversity and representativeness of Gram-negative and Gram-positive
species that were used to assess the effectiveness of different antimicrobial therapies in G. mellonella model. The data were obtained after a bibliographic search
conducted in July 2020 at PubMed (MEDLINE database) with the keyword ‘Galleria mellonella’, and subsequent individual assessment of the nature of each scientific
article.

infected with S. aureus, this natural compound increased larval Another effective approach to control bacterial infections
survival, while decreasing melanization, and bacterial load in is therapy with bacteriophages. Due to their highly specific
the hemolymph. antimicrobial properties, lytic bacteriophages have re-emerged
In another study with S. aureus, the antimicrobial potential as a promising tool for the treatment of drug-resistant bacte-
of the polyphenol epigallocatechin gallate (EGCG) was tested by rial infections. Different phages or phage cocktails have been
Knidel et al. (2019) against clinical strains with different genetic used, in treatments with single or multiple doses, to success-
profiles, level of virulence and antimicrobial susceptibilities. fully treat G. mellonella infected with A. baumannii, Enterobacter
EGCG significantly reduced the mortality of G. mellonella larvae cloacae, E. coli, K. pneumonia, P. aeruginosa and S. aureus (Forti et al.
infected with S. aureus, thus being a promising substance for the 2018; Manohar, Nachimuthu and Lopes 2018; Thiry et al. 2019;
treatment of infections caused by this agent. Tkhilaishvili et al. 2020).
Pereira et al. 11

Manohar, Nachimuthu and Lopes (2018) evaluated the ther- outlets whereby researchers can purchase specific genotypes of
apeutic potential of three different phages against strains of G. mellonella, where bred under standard conditions, is a great
E. coli, K. pneumoniae and E. cloacae using G. mellonella. Larvae limitation. Currently, there are only a few companies that sup-
infected with E. coli and E. cloacae had to be treated with three ply standardized G. mellonella larvae for scientific purposes (e.g.
phage doses at a 6 h interval to achieve a 100% survival rate. BioSystems Technology, UK). Most of the suppliers of G. mel-
However, in the case of K. pneumoniae, a single phage dose treat- lonella used in research are independent breeders specialized in
ment was sufficient. More recently, Thiry et al. (2019) showed the selling larvae as animal food rather than for use in scientific
effectiveness of new bacteriophages against emerging ST23 and research. Therefore, there is no appropriate standardization in
ST258 lineages of K. pneumoniae, reducing the mortality rate of the protocols for rearing the insects. In addition, companies that
G. mellonella larvae from 70% to 20%. supply G. mellonella for non-scientific purposes often increase
Bacteriophage therapy can also be a great strategy for the productivity by using antibiotics and juvenilizing agents that
treatment of bacterial infections associated with biofilms, such alter larval metabolism (Cutuli et al. 2019).
as those caused by S. aureus. In this sense, Tkhilaishvili et al. Some laboratories, like ours, rear their own G. mellonella for
(2020) evaluated the ability of two commercially available Staphy- scientific research. By doing so, it becomes clearer how impor-
lococcal bacteriophages (PYO and Sb) to disrupt biofilms, control tant it is to standardize the rearing protocol for G. mellonella,
biofilm formation and to control the development of an infec- and how sensitive the larvae can be to diet and environmen-
tion caused by methicillin-resistant S. aureus (MRSA). In the face tal variations, such as temperature, exposure to light, physical
of a systemic infection, both bacteriophages increased the sur- stress, humidity and proximity to moths or pupae. In this sense,
vival of G. mellonella, by either preventing or treating the infec- Jorjão et al. (2018) tested different artificial diets and observed
tion, similarly to the effect observed in the treatment with van- several physiological variations in G. mellonella. The study then
comycin. suggests a diet composition that benefits the life cycle of G.
The main advantages offered by G. mellonella in the study of mellonella, increasing the larval weight, and also stimulating
new antimicrobial strategies are: (i) precision of the inoculum their immune system by increasing the hemolymph volume
injected directly into the hemocoel, ensuring precise concentra- and hemocyte concentration, generating larvae that are more
tions of the compounds tested; (ii) the relatively large size of the resistant to infection by different microorganisms. Therefore, we
larvae, which allows more than one inoculation to be carried out emphasize that the standardization of an artificial diet, which
without major physical trauma; and (iii) it facilitates the screen- guarantees that G. mellonella are healthy physiologically and in
ing of a set of drugs, so that a smaller number of compounds terms of immune function, is important to minimize external
need to be evaluated in additional tests with vertebrates. influences on the results.
The use of G. mellonella to evaluate new antimicrobial strate- Temperature has a great influence on both G. mellonella devel-
gies is undoubtedly very important for rapid identification of opment and immune response (Wojda 2017). Studies show that
novel lead compounds, and new therapeutic strategies. How- the exposure of larvae to heat induces immune responses,
ever, there are considerations required when carrying out such increasing the expression of AMPs, while prolonged exposure
experiments in G. mellonella. The tests must be carried out with to low temperatures decreases the density of hemocytes and
bacterial doses that do not kill the larva quickly, and the tox- proteins associated with metabolic pathways and prophenolox-
icity of the compound used must also be tested. Experiments idase, affecting the larva’s susceptibility to microbial pathogens
must be organized and precisely timed so that there are no vari- (Browne et al. 2015; Wojda 2017).
ations in treatment times, especially in experiments with many Therefore, it is evident that standard G. mellonella rearing
treatments. Much attention should also be paid to the inocula- protocols, regarding diet, temperature and other factors, facil-
tion site; to avoid further trauma, it is suggested to alternate the itates reproducibility of results both within and between lab-
inoculation sides. oratories. The act of purchasing larvae from non-specialized
companies keeps the researcher from controlling situations that
might influence studies’ outcomes.
PRACTICAL CONSIDERATIONS ON THE USE OF
THE G. MELLONELLA IN RESEARCH WITH Other factors influencing reproducibility
BACTERIAL PATHOGENS
In addition to the aforementioned considerations related to
Given what has been discussed so far, the various applications the rearing of G. mellonella, other factors that influence the
and advantages offered by the G. mellonella model to scientific results obtained with infection models are worth mentioning.
research are clear. The model allows advances in research with Pre-treatments are one of those factors and can drastically alter
a wide diversity of bacterial pathogens, both in aspects related to the results of a study with G. mellonella.
virulence and to the use of new antimicrobial therapies (Figures A widely used pre-treatment is the incubation of the larvae at
S1 and S2, Supporting Information). However, beyond the critical low temperatures from one to several days. As previously men-
sense of the researcher for the interpretation of the data, taking tioned, this practice reduces the immune response of G. mel-
into account the particularities of the model and the microor- lonella and thus, the virulence of a microorganism tested can be
ganism, the success of the study and consistency of the data overestimated. However, this pre-treatment can be practiced if
obtained depend on simple factors that are often overlooked. the study involves pathogens with relatively low virulence, as
described by Browne et al. (2015).
Another common practice is leaving larvae without food for a
How is the G. mellonella that you use reared?
variable period before the experiment (Ramarao, Nielsen-Leroux
One of the greatest difficulties in standardizing experiments and Lereclus 2012; Andrea, Krogfelt and Jenssen 2019). Similar
with G. mellonella is related to the origin of the larvae used. Lar- to incubation at low temperatures, starvation also compromises
vae can be reared in research laboratories at low cost, but can the immune response of larvae making them more susceptible
also be easily purchased from outside suppliers. The lack of to infection (Banville, Browne and Kavanagh 2012).
12 Pathogens and Disease, 2020, Vol. 00, No. 00

Other factors that can influence the results are: (i) prepa- ACKNOWLEDGMENTS
ration of the inoculum (concentration of microorganisms and
The authors thank Prof. Paul Richard Langford from the Imperial
growth phase must be well defined); (ii) form of inoculation
College London (UK) for his critical analysis of this review and
(intrahemocoelic injection, the most used, or oral gavage of
valuable suggestions.
gut pathogens); (iii) definition of larval death (some researches
observe melanization, others also observe larval movement
and/or response to touch); and (iv) number of larvae used in the
SUPPLEMENTARY DATA
experiment.
One of the great advantages of using an invertebrate model is Supplementary data are available at FEMSPD online.
the possibility of working with large numbers of individuals in
parallel, and the possibility of greater statistical validity. Stud-
ies often use from 5 to 15 larvae per group (Pereira et al. 2015; FUNDING
Li et al. 2020; Russo and Macdonald 2020). Most researchers use The authors thank Conselho Nacional de Desenvolvimento
groups of 10 larvae for each treatment, with three experimen- Cientı́fico e Tecnológico—CNPq (Process n. 432065/2018-0),
tal and three biological replicates. Experiments with a smaller Coordenação de Aperfeiçoamento de Pessoal de Nı́vel Supe-
number of larvae might not be reproducible, and generate uncer- rior/Programa de Excelência Acadêmica—Finance Code 001
tainties in the interpretation of the results. (CAPES ProEx grant 23038.002486/2018-26) and Fundação de
The aforementioned experimental variables can lead to con- Amparo à Pesquisa do Estado de Minas Gerais—FAPEMIG for the
flicting results obtained with the same reference strains but car- financial support.
ried out by different research laboratories. An example is the
comparison of the results of the study by Olsen et al. (2011) to Conflict of Interest. None declared.
that of Loh et al. (2013) with the Streptococcus pyogenes strain
MGAS315. Both infected larvae with 106 CFU of bacteria, but
REFERENCES
while the first study observed 45% survival after 24 h and 25%
survival after 96 h, the second study observed a survival of 90% Aathmanathan VS, Jothi N, Prajapati VK et al. Investigation of
and 70%, respectively, at the same time points. However, each immunogenic properties of hemolin from silkworm, Bombyx
group had purchased their larvae from different suppliers and mori as carrier protein: an immunoinformatic approach. Sci
maintained the insects in different conditions. Rep 2018;8:6957.
Recently, conflicting results have also been obtained by Ambrosi C, Sarshar M, Aprea MR et al. Colonic adenoma-
Jønsson et al. (2017) and Guerrieri et al. (2019) in studies with the associated Escherichia coli express specific phenotypes.
EAEC strain 042. In the first case, 100% of G. mellonella larvae were Microbes Infect 2019;21:305–12.
killed at 24 h post-infection with an inoculum of 104 CFU/larva, Andrea A, Krogfelt KA, Jenssen H. Methods and challenges of
whereas in the second study only 60% of the larvae were dead using the greater wax moth (Galleria mellonella) as a model
at 24 h post-infection with the same strain at a higher dose (105 organism in antimicrobial compound discovery. Microorgan-
CFU/larva). In this case, the difference in the virulence profile of isms 2019;7:85.
strain 042 can also be attributed to different G. mellonella geno- Arteaga Blanco LA, Crispim JS, Fernandes KM et al. Differential
types and rearing protocols, or variations in their experimental cellular immune response of Galleria mellonella to Actinobacil-
protocols. lus pleuropneumoniae. Cell Tissue Res 2017;370:153–68.
Banville N, Browne N, Kavanagh K. Effect of nutrient deprivation
on the susceptibility of Galleria mellonella larvae to infection.
Virulence 2012;3:497–503.
Barbaro ADL, Gariboldi MB, Mastore M et al. In vivo effects of a
pro-PO system inhibitor on the phagocytosis of Xenorhabdus
nematophila in Galleria mellonella larvae. Insects 2019;10:1–19.
CONCLUSION
Betts JW, Hornsey M, Wareham DW et al. In vitro and in
The numerous advantages of the use of G. mellonella have led vivo activity of theaflavin–epicatechin combinations versus
to its widespread use as a model to study microbial pathogens. multidrug-resistant Acinetobacter baumannii. Infect Dis Ther
However, many variables can affect the outcome of such stud- 2017;6:435–42.
ies, and the interpretation of results, e.g. the source of the insect Betts JW, Phee LM, Hornsey M et al. In vitro and in vivo activ-
(purchased or laboratory-reared), and prior pre-treatment (diet, ities of tigecycline–colistin combination therapies against
temperature etc.). Microbial virulence is often complex and mul- carbapenem-resistant Enterobacteriaceae. Antimicrob Agents
tifactorial, and sometimes G. mellonella may not be an appropri- Chemother 2014;58:3541–6.
ate host for specific bacterial pathogens. Although this does not Brochado AR, Telzerow A, Bobonis J et al. Species-specific activity
invalidate the model, it reinforces that well-established ques- of antibacterial drug combinations. Nature 2018;559:259–63.
tions and protocols are pivotal to the success of experimen- Brown SE, Howard A, Kasprzak AB et al. A peptidomics study
tation with G. mellonella. After a decade-long experience with reveals the impressive antimicrobial peptide arsenal of
the model for the study of several bacterial pathogens, we have the wax moth Galleria mellonella. Insect Biochem Mol Biol
observed that the responses for each microorganism vary, and it 2009;39:792–800.
is extremely important that researchers understand the genetic Browne N, Heelan M, Kavanagh K. An analysis of the structural
background and physiology of the microorganism studied, so and functional similarities of insect hemocytes and mam-
that the correct interpretation of the results can be made. How- malian phagocytes. Virulence 2013;4:597–603.
ever, it is unquestionable that G. mellonella is a versatile, suitable Browne N, Surlis C, Maher A et al. Prolonged pre-incubation
and reliable model to conduct studies with bacterial pathogens increases the susceptibility of Galleria mellonella larvae to bac-
and antimicrobial agents. terial and fungal infection. Virulence 2015;6:458–65.
Pereira et al. 13

Brust FR, Boff L, Trentin DS et al. Macrocolony of NDM-1 pro- Guerrieri CG, Pereira MF, Galdino ACM et al. Typical and atypi-
ducing Enterobacter hormaechei subsp. oharae generates sub- cal enteroaggregative Escherichia coli are both virulent in the
populations with different features regarding the response Galleria mellonella model. Front Microbiol 2019;10:01791.
of antimicrobial agents and biofilm formation. Pathogens Haddad O, Merghni A, Elargoubi A et al. Comparative study of
2019;8:8020049 virulence factors among methicillin resistant Staphylococcus
Catalán-Nájera JC, Garza-Ramos U, Barrios-Camacho H. Hyper- aureus clinical isolates. BMC Infect Dis 2018;18:560.
virulence and hypermucoviscosity: two different but com- Hillyer JF. Insect immunology and hematopoiesis. Dev Comp
plementary Klebsiella spp. phenotypes? Virulence 2017;8:1111– Immunol 2016;58:102–18.
23. Hubrecht RC, Carter E. The 3Rs and humane experimental tech-
CDC. Antibiotic resistance threats in the United States, Technical nique: implementing change. Animals 2019;9:754.
report. U.S. Department of Health and Human Services, CDC. Idowu T, Ammeter D, Arthur G et al. Potentiation of β-lactam
2019, 1–150. antibiotics and β-lactam/β-lactamase inhibitor combina-
Chibebe J, Jr, Fuchs BB, Sabino CP et al. Photodynamic and antibi- tions against MDR and XDR Pseudomonas aeruginosa using
otic therapy impair the pathogenesis of Enterococcus faecium non-ribosomal tobramycin–cyclam conjugates. J Antimicrob
in a whole animal insect model. PLoS One 2013;8:e55926. Chemother 2019;74:2640–8.
Chung JH, Bhat A, Kim CJ et al. Combination therapy with Insua JL, Llobet E, Moranta D et al. Modeling Klebsiella pneumoniae
polymyxin B and netropsin against clinical isolates pathogenesis by infection of the wax moth Galleria mellonella.
of multidrug-resistant Acinetobacter baumannii. Sci Rep Infect Immun 2013;81:3552–65.
2016;6:28168. Jensen BH, Olsen KEP, Struve C et al. Epidemiology and clini-
Chusri S, Chongsuvivatwong V, Silpapojakul K et al. Clinical cal manifestations of enteroaggregative Escherichia coli. Clin
characteristics and outcomes of community and hospital- Microbiol Rev 2014;27:614–30.
acquired Acinetobacter baumannii bacteremia. J Microbiol Jeon J, Park JH, Yong D. Efficacy of bacteriophage treatment
Immunol Infect 2019;52:796–806. against carbapenem-resistant Acinetobacter baumannii in Gal-
Cook SM, McArthur JD. Developing Galleria mellonella as a model leria mellonella larvae and a mouse model of acute pneumo-
host for human pathogens. Virulence 2013;4:350–3. nia. BMC Microbiol 2019;19:70.
Cools F, Torfs E, Aizawa J et al. Optimization and characterization Jønsson R, Struve C, Jenssen H et al. The wax moth Galleria mel-
of a Galleria mellonella larval infection model for virulence lonella as a novel model system to study enteroaggregative
studies and the evaluation of therapeutics against Streptococ- Escherichia coli pathogenesis. Virulence 2017;8:1894–9.
cus pneumoniae. Front Microbiol 2019;10:311. Jorjão AL, Oliveira LD, Scorzoni L et al. From moths to caterpil-
Croxen MA, Finlay BB. Molecular mechanisms of Escherichia coli lars: ideal conditions for Galleria mellonella rearing for in vivo
pathogenicity. Nat Rev Microbiol 2010;8:26–38. microbiological studies. Virulence 2018;9:383–9.
Cutuli MA, Petronio Petronio G, Vergalito F et al. Galleria mellonella Kaszab E, Radó J, Kriszt B et al. Groundwater, soil and compost,
as a consolidated in vivo model hosts: new developments as possible sources of virulent and antibiotic-resistant Pseu-
in antibacterial strategies and novel drug testing. Virulence domonas aeruginosa. Int J Environ Health Res 2019;18:1–13.
2019;10:527–41. Kavanagh K, Reeves EP. Exploiting the potential of insects for
Da Silva CA, Chalouni C, Williams A et al. Chitin is a size- in vivo pathogenicity testing of microbial pathogens. FEMS
dependent regulator of macrophage TNF and IL-10 produc- Microbiol Rev 2004;28:101–12.
tion. J Immunol 2009;182:3573–82. Kavanagh K, Sheehan G. The use of Galleria mellonella larvae to
Dayan GH, Mohamed N, Scully IL et al. Staphylococcus aureus: the identify novel antimicrobial agents against fungal species of
current state of disease, pathophysiology and strategies for medical interest. J Fungi 2018;4:113.
prevention. Expert Rev Vaccines 2016;15:1373–92. Kay S, Edwards J, Brown J et al. Galleria mellonella infection model
De la Cruz MA, Morgan JK, Ares MA et al. The two-component identifies both high and low lethality of Clostridium perfrin-
system CpxRA negatively regulates the locus of enterocyte gens toxigenic strains and their response to antimicrobials.
effacement of enterohemorrhagic Escherichia coli involving Front Microbiol 2019;10:1281.
σ 32 and Lon protease. Front Cell Infect Microbiol 2016;6:11. Khairy RM, Mohamed ES, Ghany HMA et al. Phylogenic clas-
Diard M, Hardt WD. Evolution of bacterial virulence. FEMS Micro- sification and virulence genes profiles of uropathogenic E.
biol Rev 2017;41:679–97. coli and diarrhegenic E. coli strains isolated from community
Eleftherianos I, Revenis C. Role and importance of phenoloxi- acquired infections. PLoS One 2019;14:e0222441.
dase in insect hemostasis. J Innate Immun 2011;3:28–33. Kim CH, Shin YP, Noh MY et al. An insect multiligand recogni-
Esposito EP, Cervoni M, Bernardo M et al. Molecular epidemiology tion protein functions as an opsonin for the phagocytosis of
and virulence profiles of colistin-resistant Klebsiella pneumo- microorganisms. J Biol Chem 2010;285:25243–50.
niae blood isolates from the hospital agency “Ospedale dei King JG, Hillyer JF. Infection-induced interaction between the
Colli,” Naples, Italy. Front Microbiol 2018;9:01463. mosquito circulatory and immune systems. PLoS Pathog
Forti F, Roach DR, Cafora M et al. Design of a broad-range 2012;8:e1003058.
bacteriophage cocktail that reduces Pseudomonas aeruginosa Klein K, Sonnabend MS, Frank L et al. Deprivation of the periplas-
biofilms and treats acute infections in two animal models. mic chaperone SurA reduces virulence and restores antibi-
Antimicrob Agents Chemother 2018;62:e02573–17. otic susceptibility of multidrug-resistant Pseudomonas aerug-
Graf AC, Leonard A, Schäuble M et al. Virulence factors pro- inosa. Front Microbiol 2019;10:00100.
duced by Staphylococcus aureus biofilms have a moonlighting Klein RD, Hultgren SJ. Urinary tract infections: microbial patho-
function contributing to biofilm integrity. Mol Cell Proteomics genesis, host–pathogen interactions and new treatment
2019;18:1036–53. strategies. Nat Rev Microbiol 2020;18:211–26.
Graham ML, Prescott MJ. The multifactorial role of the 3Rs in Knidel C, Pereira MF, Barcelos DHF et al. Epigallocatechin gal-
shifting the harm-benefit analysis in animal models of dis- late has antibacterial and antibiofilm activity in methicillin
ease. Eur J Pharmacol 2015;759:19–29. resistant and susceptible Staphylococcus aureus of different
14 Pathogens and Disease, 2020, Vol. 00, No. 00

lineages in non-cytotoxic concentrations. Nat Prod Res 2019; Morin N, Santiago AE, Ernst RK et al. Characterization of the AggR
33:1698575. regulon in enteroaggregative Escherichia coli. Infect Immun
Laabei M, Uhlemann AC, Lowy FD et al. Evolutionary trade-offs 2013;81:122–32.
underlie the multi-faceted virulence of Staphylococcus aureus. Mukherjee K, Amsel D, Kalsy M et al. MicroRNAs regulate
PLoS Biol 2015;13:1002229. innate immunity against uropathogenic and commensal-
Lam MMC, Seemann T, Tobias NJ et al. Comparative analysis of like Escherichia coli infections in the surrogate insect model
the complete genome of an epidemic hospital sequence type Galleria mellonella. Sci Rep 2020;10:2570.
203 clone of vancomycin-resistant Enterococcus faecium. BMC Nataro JP, Kaper JB. Diarrheagenic Escherichia coli. Clin Microbiol
Genomics 2013;14:595. Rev 1998;11:142–201.
Lee AS, De Lencastre H, Garau J et al. Methicillin-resistant Staphy- Nath S, Moussavi F, Abraham D et al. In vitro and in vivo activity of
lococcus aureus. Nat Rev Dis Prim 2018;4:18033. single and dual antimicrobial agents against KPC-producing
Leshkasheli L, Kutateladze M, Balarjishvili N et al. Efficacy of Klebsiella pneumoniae. J Antimicrob Chemother 2018;73:431–6.
newly isolated and highly potent bacteriophages in a mouse Nathan C. Resisting antimicrobial resistance. Nat Rev Microbiol
model of extensively drug-resistant Acinetobacter baumannii 2020;18:259–60.
bacteraemia. J Glob Antimicrob Resist 2019;19:255–61. Niu T, Guo L, Luo Q et al. Wza gene knockout decreases Acineto-
Li G, Shi J, Zhao Y et al. Identification of hypervirulent Kleb- bacter baumannii virulence and affects Wzy-dependent cap-
siella pneumoniae isolates using the string test in combination sular polysaccharide synthesis. Virulence 2020;11:1–13.
with Galleria mellonella infectivity. Eur J Clin Microbiol Infect Dis Okeke IN, Lamikanra A, Czeczulin J et al. Heterogeneous vir-
2020:03890. ulence of enteroaggregative Escherichia coli strains isolated
Li L, Chen H, Liu Y et al. Synergistic effect of linezolid with fos- from children in southwest Nigeria. J Infect Dis 2000;181:252–
fomycin against Staphylococcus aureus in vitro and in an exper- 60.
imental Galleria mellonella model. J Microbiol Immunol Infect Okhuysen PC, DuPont HL. Enteroaggregative Escherichia coli
2019;12:007. (EAEC): a cause of acute and persistent diarrhea of worldwide
Loh JMS, Adenwalla N, Wiles S et al. Galleria mellonella larvae importance. J Infect Dis 2010;202:503–5.
as an infection model for group A streptococcus. Virulence Oliveira D, Borges A, Simões M. Staphylococcus aureus toxins
2013;4:419–28. and their molecular activity in infectious diseases. Toxins
López Hernández Y, Yero D, Pinos-Rodrı́guez JM et al. Animals 2018;10:252.
devoid of pulmonary system as infection models in the study Olsen RJ, Ebru Watkins M, Cantu CC et al. Virulence of serotype
of lung bacterial pathogens. Front Microbiol 2015;6:38. M3 group a streptococcus strains in wax worms (Galleria mel-
Lo Sciuto A, Cervoni M, Stefanelli R et al. Genetic basis and physi- lonella larvae). Virulence 2011;2:111–9.
ological effects of lipid a hydroxylation in Pseudomonas aerug- Pati NB, Doijad SP, Schultze T et al. Enterobacter bugandensis: a
inosa PAO1. Pathogens 2019;8:291. novel enterobacterial species associated with severe clinical
Lu A, Zhang Q, Zhang J et al. Insect prophenoloxidase: the view infection. Sci Rep 2018;8:5392.
beyond immunity. Front Physiol 2014;5:252. Pereira MF, Rossi CC, de Queiroz MV et al. Galleria mellonella is
Ma D, Mandell JB, Donegan NP et al. The toxin-antitoxin MazEF an effective model to study Actinobacillus pleuropneumoniae
drives Staphylococcus aureus biofilm formation, antibiotic tol- infection. Microbiology 2015;161:387–400.
erance, and chronic infection. mBio 2019;10:e01658–19. Pereira MF, Rossi CC, Seide LE et al. Antimicrobial resistance,
Manohar P, Nachimuthu R, Lopes BS. The therapeutic poten- biofilm formation and virulence reveal Actinobacillus pleu-
tial of bacteriophages targeting Gram-negative bacteria ropneumoniae strains’ pathogenicity complexity. Res Vet Sci
using Galleria mellonella infection model. BMC Microbiol 2018;118:498–501.
2018;18:1234. Pérez-Montarelo D, Viedma E, Murcia M et al. Pathogenic char-
Maravilla E, Le DP, Tran JJ et al. Apolipophorin III interac- acteristics of Staphylococcus aureus endovascular infection
tion with phosphatidylglycerol and lipopolysaccharide. Chem isolates from different clonal complexes. Front Microbiol
Phys Lipids 2020;229:104909. 2017;8:917.
Medina-Rojas M, Stribling W, Snesrud E et al. Comparison of Petrosillo, Taglietti, Granata. Treatment options for colistin resis-
Pseudomonas aeruginosa strains reveals that exolysin A toxin tant Klebsiella pneumoniae: present and future. J Clin Med
plays an additive role in virulence. Pathog Dis 2020;78:ftaa010. 2019;8:934.
Meir M, Grosfeld T, Barkan D. Establishment and validation Pitout JDD, Nordmann P, Poirel L. Carbapenemase-producing
of Galleria mellonella as a novel model organism to study Klebsiella pneumoniae, a key pathogen set for global nosoco-
Mycobacterium abscessus infection, pathogenesis, and treat- mial dominance. Antimicrob Agents Chemother 2015;59:5873–
ment. Antimicrob Agents Chemother 2018,62:e02539–17. 84.
Mills G, Dumigan A, Kidd T et al. Identification and char- Poolman JT. Escherichia coli. 2nd edn. New York: Elsevier, 2017.
acterization of two Klebsiella pneumoniae lpxL lipid A late Propst CN, Pylypko SL, Blower RJ et al. Francisella philomiragia
acyltransferases and their role in virulence. Infect Immun infection and lethality in mammalian tissue culture cell
2017;85:e00068–17. models, Galleria mellonella, and BALB/c mice. Front Microbiol
Minrovic BM, Jung D, Melander RJ et al. A new class of adjuvants 2016;7:696.
enables lower dosing of colistin against Acinetobacter bauman- Ramarao N, Nielsen-Leroux C, Lereclus D. The insect Galleria mel-
nii. ACS Infect Dis 2018;176:139–48. lonella as a powerful infection model to investigate bacterial
Moghaddam MRB, Tonk M, Schreiber C et al. The potential of pathogenesis. J Vis Exp 2012;70:e4392.
the Galleria mellonella innate immune system is maximized Russo TA, MacDonald U. The Galleria mellonella infection model
by the co-presentation of diverse antimicrobial peptides. Biol does not accurately differentiate between hypervirulent and
Chem 2016;397:939–45 classical Klebsiella pneumoniae. mSphere 2020;5:e00850–19.
Russo TA, Marr CM. Hypervirulent Klebsiella pneumoniae. Clin
Microbiol Infect 2019;32:e00001–19.
Pereira et al. 15

Salze M, Muller C, Bernay B et al. Study of key RNA metabolism Twittenhoff C, Heroven AK, Mühlen S et al. An RNA thermometer
proteins in Enterococcus faecalis. RNA Biol 2020;17:794–804. dictates production of a secreted bacterial toxin. PLoS Pathog
Sarantuya J, Nishi J, Wakimoto N et al. Typical enteroaggregative 2020;16:e1008184.
Escherichia coli is the most prevalent pathotype among E. coli Vergis J, Malik SS, Pathak R et al. Antimicrobial efficacy of
strains causing diarrhea in Mongolian children. J Clin Micro- indolicidin against multi-drug resistant enteroaggregative
biol 2004;42:133–9. Escherichia coli in a Galleria mellonella model. Front Microbiol
Schwidder M, Heinisch L, Schmidt H. Genetics, toxicity, and 2019;10:2723.
distribution of enterohemorrhagic Escherichia coli hemolysin. Wand ME, McCowen JWI, Nugent PG et al. Complex interac-
Toxins 2019;11:502. tions of Klebsiella pneumoniae with the host immune sys-
Seed KD, Dennis JJ. Development of Galleria mellonella as an alter- tem in a Galleria mellonella infection model. J Med Microbiol
native infection model for the Burkholderia cepacia complex. 2013;62:1790–8.
Infect Immun 2008;76.3:1267–75. Wang Q, Ren M, Liu X et al. Peptidoglycan recognition proteins in
Shaik HA, Sehnal F. Hemolin expression in the silk glands of Gal- insect immunity. Mol Immunol 2019;106:69–76.
leria mellonella in response to bacterial challenge and prior to Weidensdorfer M, Ishikawa M, Hori K et al. The Acinetobacter
cell disintegration. J Insect Physiol 2009;55:781–7. trimeric autotransporter adhesin Ata controls key virulence
Sharma-Kuinkel BK, Mongodin EF, Myers JR et al. Potential influ- traits of Acinetobacter baumannii. Virulence 2019;10:68–81.
ence of Staphylococcus aureus clonal complex 30 genotype Weigel WA, Dersch P. Phenotypic heterogeneity: a bacterial vir-
and transcriptome on hematogenous infections. Open Forum ulence strategy. Microbes Infect 2018;20:570–7.
Infect Dis 2017;2:2633851. Williamson DA, Mills G, Johnson JR et al. In vivo correlates
Sheehan G, Dixon A, Kavanagh K. Utilization of Galleria mel- of molecularly inferred virulence among extraintestinal
lonella larvae to characterize the development of Staphylococ- pathogenic Escherichia coli (ExPEC) in the wax moth Galleria
cus aureus infection. Microbiology 2019;165:863–75. mellonella model system. Virulence 2014;5:388–93.
Sheehan G, Garvey A, Croke M et al. Innate humoral immune Wojda I. Immunity of the greater wax moth Galleria mellonella.
defences in mammals and insects: the same, with differ- Insect Sci 2017;24:342–57.
ences? Virulence 2018;9:1625–39. World Health Organization. List of bacteria for which new antibi-
Sheehan G, Konings M, Lim W et al. Proteomic analysis of the otics are urgently needed. 2017. Switzerland: W.H.O. Press.
processes leading to Madurella mycetomatis grain formation in Wu G, Liu Y, Ding Y et al. Ultrastructural and functional char-
Galleria mellonella larvae. PLoS Negl Trop Dis 2020;14:e0008190. acterization of circulating hemocytes from Galleria mellonella
Shi Q, Lan P, Huang D et al. Diversity of virulence level phe- larva: cell types and their role in the innate immunity. Tissue
notype of hypervirulent Klebsiella pneumoniae from different Cell 2016;48:297–304.
sequence type lineage. BMC Microbiol 2018;18:1236. Wyres KL, Wick RR, Judd LM et al. Distinct evolutionary dynam-
Spano LC, da Cunha KF, Monfardini MV et al. High prevalence of ics of horizontal gene transfer in drug resistant and virulent
diarrheagenic Escherichia coli carrying toxin-encoding genes clones of Klebsiella pneumoniae. PLos Genet 2019;15:e1008114.
isolated from children and adults in southeastern Brazil. Yang H, Chen G, Hu L et al. Enhanced efficacy of imipenem-
BMC Infect Dis 2017;17:773. colistin combination therapy against multiple-drug-
Stączek S, Zdybicka-Barabas A, Mak P et al. Studies on localiza- resistant Enterobacter cloacae: in vitro activity and a Galleria
tion and protein ligands of Galleria mellonella apolipophorin mellonella model. J Microbiol Immunol Infect 2016;51:70–5.
III during immune response against different pathogens. J Yuan Y, Zai Y, Xi X et al. A novel membrane-disruptive antimi-
Insect Physiol 2018;105:18–27. crobial peptide from frog skin secretion against cystic fibro-
Tam K, Torres VJ. Staphylococcus aureus secreted toxins and extra- sis isolates and evaluation of anti-MRSA effect using Galleria
cellular enzymes. Microbiol Spectr 2019;7:GPP3–0039-2018. mellonella model. Biochim Biophys Acta Gen Subj 2019;1863:849–
Thieme L, Hartung A, Makarewicz O et al. In vivo synergism of 56.
ampicillin, gentamicin, ceftaroline and ceftriaxone against Zhao X, Palma Medina LM, Stobernack T et al. Exoproteome het-
Enterococcus faecalis assessed in the Galleria mellonella infec- erogeneity among closely related Staphylococcus aureus t437
tion model. J Antimicrob Chemother 2020; 75:2173–81. isolates and possible implications for virulence. J Proteome
Thiry D, Passet V, Danis-wlodarczyk K et al. New bacteriophages Res 2019;18:2859–74.
against emerging lineages ST23 and ST258 of Klebsiella pneu- Zheng J, Wu Y, Lin Z et al. ClpP participates in stress tolerance,
moniae and efficacy assessment in Galleria mellonella larvae. biofilm formation, antimicrobial tolerance, and virulence of
Viruses 2019;11:411. Enterococcus faecalis. BMC Microbiol 2020;20:1–18.
Tkhilaishvili T, Wang L, Tavanti A et al. Antibacterial effi- Zheng Z, Tharmalingam N, Liu Q et al. Synergistic efficacy of
cacy of two commercially available bacteriophage formula- Aedes aegypti antimicrobial peptide cecropin A2 and tetra-
tions, Staphylococcal bacteriophage and PYO bacteriophage, cycline against Pseudomonas aeruginosa. Antimicrob Agents
against methicillin-resistant Staphylococcus aureus: preven- Chemother 2017;61:e00686–17.
tion and eradication of biofilm formation and control of a Zhou H, Larkin PMK, Huang J et al. Discovery of a novel hypervir-
systemic infection of Galleria mellonella larvae. Front Microbiol ulent Acinetobacter baumannii strain in a case of community-
2020;11:110. acquired pneumonia. Infect Drug Resist 2020;13:1147–53.
Tsai CJY, Loh JMS, Proft T. Galleria mellonella infection models Ziebandt AK, Kusch H, Degner M et al. Proteomics uncovers
for the study of bacterial diseases and for antimicrobial drug extreme heterogeneity in the Staphylococcus aureus exopro-
testing. Virulence 2016;7:214–29. teome due to genomic plasticity and variant gene regulation.
Turner NA, Sharma-Kuinkel BK, Maskarinec SA et al. Methicillin- Proteomics 2010;10:1634–44.
resistant Staphylococcus aureus: an overview of basic and clin-
ical research. Nat Rev Microbiol 2019;17:203–18.

You might also like