ArticlePDF AvailableLiterature Review

Galleria mellonella—A Model for the Study of aPDT—Prospects and Drawbacks

Authors:

Abstract and Figures

Galleria mellonella is a promising in vivo model insect used for microbiological, medical, and pharmacological research. It provides a platform for testing the biocompatibility of various compounds and the kinetics of survival after an infection followed by subsequent treatment, and for the evaluation of various parameters during treatment, including the host–pathogen interaction. There are some similarities in the development of pathologies with mammals. However, a limitation is the lack of adaptive immune response. Antimicrobial photodynamic therapy (aPDT) is an alternative approach for combating microbial infections, including biofilm-associated ones. aPDT is effective against Gram-positive and Gram-negative bacteria, viruses, fungi, and parasites, regardless of whether they are resistant to conventional treatment. The main idea of this comprehensive review was to collect information on the use of G. mellonella in aPDT. It provides a collection of references published in the last 10 years from this area of research, complemented by some practical experiences of the authors of this review. Additionally, the review summarizes in brief information on the G. mellonella model, its advantages and methods used in the processing of material from these larvae, as well as basic knowledge of the principles of aPDT.
Content may be subject to copyright.
Citation: Bugyna, L.; Kendra, S.;
Bujdáková, H. Galleria mellonella—A
Model for the Study of aPDT—
Prospects and Drawbacks.
Microorganisms 2023,11, 1455.
https://doi.org/10.3390/
microorganisms11061455
Academic Editors: Giulio Petronio
Petronio, Roberto Di Marco and
Laura Pietrangelo
Received: 30 April 2023
Revised: 29 May 2023
Accepted: 30 May 2023
Published: 31 May 2023
Copyright: © 2023 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
microorganisms
Review
Galleria mellonella—A Model for the Study of aPDT—Prospects
and Drawbacks
Larysa Bugyna , Samuel Kendra and Helena Bujdáková*
Faculty of Natural Sciences, Department of Microbiology and Virology, Comenius University in Bratislava,
Ilkovicova 6, 84215 Bratislava, Slovakia; larysa.bugyna@uniba.sk (L.B.); kendra4@uniba.sk (S.K.)
*Correspondence: helena.bujdakova@uniba.sk; Tel.: +421-2-9014-9436
Abstract:
Galleria mellonella is a promising
in vivo
model insect used for microbiological, medical,
and pharmacological research. It provides a platform for testing the biocompatibility of various
compounds and the kinetics of survival after an infection followed by subsequent treatment, and
for the evaluation of various parameters during treatment, including the host–pathogen interaction.
There are some similarities in the development of pathologies with mammals. However, a limitation is
the lack of adaptive immune response. Antimicrobial photodynamic therapy (aPDT) is an alternative
approach for combating microbial infections, including biofilm-associated ones. aPDT is effective
against Gram-positive and Gram-negative bacteria, viruses, fungi, and parasites, regardless of
whether they are resistant to conventional treatment. The main idea of this comprehensive review
was to collect information on the use of G. mellonella in aPDT. It provides a collection of references
published in the last 10 years from this area of research, complemented by some practical experiences
of the authors of this review. Additionally, the review summarizes in brief information on the G.
mellonella model, its advantages and methods used in the processing of material from these larvae, as
well as basic knowledge of the principles of aPDT.
Keywords:
Galleria mellonella; larvae infection; antimicrobial photodynamic therapy; photosensitizer;
irradiation
1. Introduction
In modern medicine and pharmaceutical research, the selection of the appropriate
choice of
in vivo
model has been critical [
1
6
]. Research involving vertebrate animals is
subject to strict rules and introduces a number of ethical problems. The European Science
Foundation promotes the need for an ethical approach to each animal experiment. In 1986,
the Council of Europe and the European Union (EU) issued guidelines and legislation on the
use of animals for scientific purposes. Several organizations have prepared guidelines for
their ethical use, and in many countries this is controlled at the level of national legislative
norms. For EU members, national legislation must meet the requirements of Council
Directive 2010/63/EU of the European Parliament, which has been updated from time to
time [
7
,
8
]. Planning any research that involves animals requires following the rules of the
“3 Rs”—replace, reduce, refine. This means that firstly, if possible, it is necessary to replace
vertebrates with invertebrates; secondly, if this is impossible, it is important to reduce their
use to a minimum; and thirdly, to refine the research in such a way as to minimize the
suffering of vertebrates. At the same time, obtaining reliable results should be ensured [
8
,
9
].
Such standards are not so strict for invertebrates, such as the nematode Caenorhabditis
elegans [
10
,
11
], the fruit fly Drosophila melanogaster [
12
,
13
], zebrafish Danio rerio [
14
,
15
], and
the wax moth larvae of Galleria mellonella. The latter is a universal invertebrate model
suitable for conducting various studies that evaluate many different parameters [
4
,
16
22
].
G. mellonella has several advantages over other non-vertebrate models (mentioned in
detail in the next section). Additionally, it has some similarities to mammals in terms of
the development of pathologies with mammals [
2
,
4
,
23
]. G. mellonella larvae have a wide
Microorganisms 2023,11, 1455. https://doi.org/10.3390/microorganisms11061455 https://www.mdpi.com/journal/microorganisms
Microorganisms 2023,11, 1455 2 of 21
applicability for each type of assay, which may inform the prospects for further
in vivo
studies in mammals. However, their limitation is a lack of adaptive immune response,
namely the elevation of antibodies and cytokines and the participation of leukocyte killer
cells and dendritic cells, which makes it difficult to predict the immune response in mam-
mals [
23
]. Another obstacle is the lack of availability of mutant larvae, which makes genetic
studies problematic [
2
]. On the other hand, the inoculation of G. mellonella larvae is rapid,
so results can be obtained within a few days. This model is suitable not only for testing
the biocompatibility of different compounds and the kinetics of survival after an infection
followed by subsequent treatment, but also for the evaluation of different parameters
during treatment, including the host–pathogen interaction [4,16,17,24,25].
Antimicrobial photodynamic inactivation (aPDI) is an alternative strategy for fighting
against microorganisms and their biofilms. This approach is based on the use of a non-toxic
dye-photosensitizer (PS), a source of light with the proper wavelength, and the presence of
oxygen. The optimal interplay of all the above factors results in oxidative stress leading to
the death of target cells [
26
,
27
]. In aPDI, there is no specifically targeted component, but
it causes general damage in the cell. Therefore, it is more difficult for microorganisms to
adapt, or even to develop resistance [28].
The main idea of this comprehensive review was to collect information on the use
of G. mellonella in the study of antimicrobial photodynamic therapy (aPDT). Moreover,
the review summarizes in brief information on the G. mellonella model, its advantages,
and the methods used in the processing of material from these larvae. The information
from published works is complemented by the experimental experiences of the authors of
this review.
2. General Characterization of G. mellonela and Significance for Microbiological Research
The larvae of G. mellonella inhabit honeybee hives and feed on bee honeycombs, where
their subsequent pupation takes place. The duration of the life cycle is 8–12 weeks, in-
cluding 5–6 weeks in the larval stage, and larvae are usually about 3 cm long [
29
34
].
The advantages of G. mellonella compared to other invertebrate experimental objects can
be summarized as follows: (a) larger larval dimension, which facilitates experimental
manipulations; (b) ability to actively grow over a wider temperature range (20
C–37
C);
(c) shorter period of data acquisition (several days compared to the weeks of other inverte-
brate experimental objects). In addition, the data acquired on G. mellonella are comparable
to the studies obtained on vertebrate animals [
2
,
35
37
] as larvae can be tested at the physi-
ological temperature of vertebrates. This is an important factor that allows the study of
temperature-dependent virulence factors [
24
,
29
,
38
43
]. In addition, this invertebrate model
is capable of reproducing the clinical signs observed in human infections [42,44,45].
Additional opportunities were identified after the G. mellonella genome was success-
fully sequenced. The level of homology between G. mellonella and humans, mice, or other
model organisms has been determined [
46
,
47
]. Lange et al. (2018) published the results
from genome sequencing using PacBio’s long-read technology. They showed that the
G. mellonella genome consists of 37 genes coding for 13 proteins, 2 rRNA, and 22 transport
RNA. These results greatly contributed to the wider use of this invertebrate model and the
replacement of vertebrates in biomedical research [4851].
Previously, there were no standardized larvae of G. mellonella, which was a significant
obstacle to its wider use. For many years, they were only commercially available as food for
reptiles and birds, so they were bred, raised, and kept under various conditions. However,
they are now bred specifically for research without the addition of antibiotics or hormones
to the feed. Their age and weight are also monitored, and the cuticle is disinfected to prevent
infections in control groups [
3
,
52
]. The most common method of infection of G. mellonella
larvae is subcutaneous microinjection. Pathogens can also be administered orally, but
commercially available larvae are in the final stage of maturation before pupation, during
which time they are almost non-feeding. In this regard, oral administration is carried out
using a special probe or at an early stage of maturation [5254].
Microorganisms 2023,11, 1455 3 of 21
G. mellonella is an excellent experimental model for the preliminary screening of the
toxicity and antimicrobial activity of various compounds and disinfectants [
3
,
5
,
55
58
]. For
example, manganese-based compounds [Mn(bpqa-
κ3
N)(CO)
3
]Br, [Mn(bqpa
κ3
N)(CO)
3
]Br,
[Mn(CO)
3
(tqa-
κ3
N)]Br, and [Mn(CO)
3
(tpa-
κ3
N)Br, which damage the integrity of the
bacterial membrane, demonstrated antibacterial properties with no toxicity to G. mel-
lonella [
59
]. Similarly, silver nanoparticles (AgNPs) tested by Thomaz et al. (2020) demon-
strated effective antimicrobial activity against Pseudomonas aeruginosa [
60
]. Larva intra-
hemocoel injections were carried out with the antimicrobial peptide (Naphthalene-2-
ly)-acetyl-diphenylalanine-dilysine-OH (NapFFKK-OH) to test its activity against gram-
positive and gram-negative microorganisms [61].
Due to the increase in resistance to antibiotics, silver impurities are widely used, from
lunch boxes to medical device implants. For example, the effectiveness of silver acetate
against the carbapenem-resistant Acinetobacter baumannii was investigated. Using this
compound, the infection of G. mellonella larvae was under control, leading to significantly
improved survival. This study also demonstrated the selective toxicity of silver acetate to
bacterial pathogens without harmful effects on larvae [
62
]. In another study, the effective-
ness of probiotics was studied. Larvae were pre-inoculated with one of two commonly
used probiotic bacteria, Lactobacillus rhamnosus GG [
63
,
64
] or Clostridium butyricum Miyairi
588 [
63
], and then challenged with Salmonella enterica Typhimurium, enteropathogenic Es-
cherichia coli, or Listeria monocytogenes [
44
,
65
,
66
]. The survival rates were increased in larvae
pre-treated with probiotics compared to the control group inoculated with pathogens alone.
Hematocyte density also increased, indicating that both probiotics evocated an immune
response [
63
]. It was also established that G. mellonella larvae can be used to assess the
virulence of anaerobic bacteria of Clostridium perfringens [
67
,
68
]. The results demonstrated
that C. perfringens infection activated the melanization pathway, leading to melanin deposi-
tion. Another study proved the effectiveness of available antibiotics against the biofilms
of multi-drug-resistant Pseudomonas aeruginosa and Klebsiella pneumoniae strains [
69
]. In
addition, the use of G. mellonella larvae makes it possible to evaluate the antibacterial
efficiency of various plant extracts and their ability to modulate the immune response. For
example, pomegranate glycolic extract was effective against Porphyromonas gingivalis, and
it prolonged larval survival compared to the untreated control [70].
In summary, G. mellonella has been used for testing many infections caused by different
gram-positive and gram-negative bacteria. Among gram-positive microorganisms, Staphylo-
coccus aureus [
71
77
], Streptococcus pyogenes [
78
80
], Streptococcus pneumoniae [
81
83
], Strepto-
coccus mutans [
19
,
20
,
84
86
], L. monocytogenes [
4
,
44
,
65
,
66
,
87
,
88
], Enterococus faecalis [
89
92
],
Enterococcus faecium [
93
96
], L. rhamnosus GG [
63
,
64
], C. butyricum Miyairi 588 [
63
], C. perfrin-
gens [
67
,
68
], Mycobacterium bovis [
23
], Mycobacterium abscessus [
97
,
98
], and Mycobacterium
tuberculosis [
99
102
] were mentioned. Among gram-negative bacteria, E. coli [
103
106
],
S. enterica Typhimurium [
107
109
], K. pneumonia [
110
113
], A. baumanii [
39
,
114
117
], Fran-
cisella tularensis [
118
120
], P. aeruginosa [
60
,
121
,
122
], and P. gingivalis [
70
,
123
,
124
] have been
involved. G. mellonella was also used for testing representatives of fungal pathogens, such
as Candida albicans [
21
,
25
,
125
128
], Candida dubliniensis [
21
,
25
,
129
,
130
], Aspergillus fumiga-
tus [
38
,
131
,
132
], Cryptococcus neoformans [
57
,
133
135
],and Madurella mycetomatis [
45
,
136
138
],
and also viruses [139142] and bacteriophages [143150].
It has already been mentioned that G. mellonella larvae are a suitable
in vivo
model for
studies related to drug safety and efficacy. Additionally, they can be used for the study of
host–pathogen interactions [
2
,
151
,
152
]. An advantage is the very good survival of G. mel-
lonella at the temperature of the human body, and that they often exhibit symptoms of the
pathogenesis of various diseases similar to those manifested in humans [
4
,
79
,
97
,
153
155
].
For example, larvae infected with streptococci manifested clear signs of invasive infection.
Specifically, these included melanization and the formation of a destructive abscess-like
lesion at the inoculation site. These abscesses consisted of dense necrotic tissue in the center
and microorganisms. They were surrounded by a band of host hemocytes, coagulated
hemolymph, and the extracellular pigment melanin. According to the authors, these fea-
Microorganisms 2023,11, 1455 4 of 21
tures are similar to the histopathology commonly seen in mouse and monkey models and
could also be compared with severe soft-tissue infections observed in humans [76].
The protection of G. mellonella from microbial infection has been under intensive study,
and there are some similarities with humans. While the cuticle mimics the skin
[151154]
,
the immune response mechanism shows signs related to the innate immunity of verte-
brates [
156
159
]. Hemocoel contains hemocytes with a similar function to human neu-
trophils that participate in phagocytosis, during which reactive oxygen species (ROS) are
generated [
160
163
]. The G. mellonella larvae enable the study of the influence of infection
on the development of oxidative stress and the antioxidant defense system. Additionally, it
has been observed that apoptosis can be initiated during infection [
164
]. Insect hemocyte
extracellular traps (IHETs) were recently described. IHETs act via hemolymph coagulation
and melanization, which contributes to the immobilization and killing of bacteria. These
processes are mediated by a significant release of hemocytes in G. mellonella [156].
Soluble effector molecules orchestrate the humoral response and include complement-
like proteins, such as melanin, and antimicrobial peptides, such as gallerimycin and
galiomicin, which protect G. mellonella from fungal infection, or cecropin, which proved to
be effective during infection with Mycobacterium bovis BCG lux [21,23,46,165167].
Several approaches are currently available, and these have been adapted and opti-
mized to study various processes in G. mellonella. This analysis could be summarized
into two main areas of study: (i) the kinetics of survival after the testing of an infec-
tion/treatment; (ii) the host–pathogen interaction. The latter scope includes quantitative
analysis, but also qualitative ones that consider hemolymph analysis. Both areas can also in-
volve some molecular biology approaches. Some procedures optimized for the G. mellonella
study are briefly mentioned below with the relevant references.
To determine the progress of infection and the effectiveness of treatment, counting
the number of dead larvae, progress in melanization, the direct counting of pathogens
in body tissues, and histology could be options for evaluation. When determining the
host–pathogen interaction, the most frequent approach is a quantitative analysis providing
information on the number of hemocytes in the hemolymph, the results of which can
give an idea of the level of the immune response [
168
171
]. For this purpose, counting the
density of hemocytes, recalculated per 1 mL of hemolymph, can also be conducted [
172
,
173
].
The viability of hemocytes can be investigated using an MTT colorimetric assay. [
174
]. To
characterize the different types of hemocytes, light or phase-contrast microscopy, Giemsa
staining, or neutral red staining is recommended. The enzymatic activity of hemolymph
can be determined by measuring the concentration of insect enzymes involved in immunity,
such as lysozyme and superoxide dismutase [
171
,
175
177
]. The label-free quantification
and untargeted analysis of the complete protein profile of hemolymph is usually performed
by proteomic analysis, or proteins can be identified via 2D electrophoresis [
23
,
175
,
178
184
].
The implementation of molecular biology is necessary to obtain more detailed informa-
tion from all the above-mentioned aspects of the study of G. mellonella. For the expression
of genes coding for antimicrobial peptides and immunity-related genes, a quantitative
RT-PCR or transcriptomic analysis is optimal [23,175,178,184186].
The
in vitro
analysis of phagocytosis is performed using fluorescent microscopy of
spotted fluorescent bacteria [
164
,
172
,
174
,
187
,
188
]. Phagocytosis in hemolymph
in vivo
is
analyzed using the same method [
189
]. A detailed study of macrophage activation is
necessary to understand the level of release of ROS or nitrogen species, as well as regula-
tory enzymes [
190
]. Macrophage activation is investigated using the Greiss assay, which
analyzes the release of active nitrogen forms [
37
]. More detailed studies of macrophage
activation are related to the study of DNA damage (ELISA), lipid peroxidation level (mal-
onic aldehyde level), catalase level (fluorometric resorufin assay), or superoxide dismutase
(ELISA) [160,190,191].
In addition to the above-mentioned conventional methods, G. mellonella is a suitable
model for the study of various aspects of aPDT. This issue is addressed in the following
Microorganisms 2023,11, 1455 5 of 21
chapter, including a table summarizing the experimental research over approximately the
last 10 years, since the first work on testing aPDT on G. mellonella was published.
3. Principles of aPDT and the Use of G. mellonella in aPDT
Photodynamic therapy (PDT) was discovered more than a century ago. Its essence
was revealed in detail by Raab, who published a study in 1900 on the use of aPDT as a
cytotoxic technique designed to treat tumors, as well as infectious pathologies [192,193].
The therapeutic effect of PDT (aPDT) is achieved using a photosensitizer (PS) that
is irradiated with light, the emission spectrum of which corresponds to the absorption
spectrum of the PS. In the presence of molecular oxygen, ROS (superoxide, hydroxyl radical,
etc.) and singlet oxygen are generated, which cause the irreversible destruction of a large
range of biomolecules, including nucleic acids, lipids, and proteins [
58
,
93
,
194
]. PDT is
already in use as an alternative approach for the control of malignant diseases. A review
by Ferreira dos Santos et al. (2019) nicely summarized [
179
] the current state of the art
in PDT research and treatment focused on cancer. The authors also introduced in detail
PSs for practical use in the treatment of many cancers. For example, Porfimer, sodium
(Photofrin) was the first PS approved by the Canadian Health Agency in 1993 for the
treatment of bladder cancer. In 1998, the U.S. Food and Drug Administration approved it
for the treatment of early-stage lung cancer. Currently, 11 additional countries in Europe
have accepted the practical use of this PS [
195
]. While the PDT practiced in cancer therapy
is developing dramatically, the application of aPDT for the eradication of pathogenic
microorganisms and viruses is still at a very early stage and has only been developing
faster in the last decade. Nevertheless, several examples of the practical application of
aPDT have already been described, mainly in the treatment of oral diseases. A clinical
study by Fonseca et al. (2022) demonstrated that aPDT reduced the number of infected
anatomical sites in patients with oral candidiasis [
196
]. Another clinical study by Shetty et
al. (2022) proved that a single session of aPDT as an adjunct to mechanical debridement
is effective at reducing peri-implant soft tissue inflammation and oral yeast colonization
in patients with peri-implant mucositis [
197
]. Alves-Silva et al. (2023) used aPDT as an
adjunct to a chemo-mechanical preparation, and it was effective at improving root canal
disinfection and reducing the lipopolysaccharide and lipoteichoic acid levels in teeth with
primary endodontic infection [198].
Generally, aPDI should be an effective method for the eradication of a wide range of
microorganisms, including both gram-positive and gram-negative bacteria, viruses, fungi,
and parasites [
26
28
,
199
201
]. Due to the fact that PDI is multi-targeted, microorganisms
are not able to develop resistance [
202
,
203
]. Moreover, PDI is highly effective against
microorganisms resistant to conventional antimicrobials [
204
208
]. For instance, Štefánek
et al. (2022) used aPDI for the eradication of Candida auris biofilms resistant to antifungal
agents. They found that aPDI significantly decreased the survival of C. auris biofilm
cells, and thus proved to have great potential for the eradication of multi-resistant yeasts.
Furthermore, the observed upregulation of the MDR1 and CDR1 genes did not affect the
overall efficacy of methylene blue-mediated aPDI on biofilms formed by C. auris clinical
isolates, regardless of their sensitivity or resistance [204].
Since aPDT is still under development, optimal models are necessary to investi-
gate not only the effectiveness of treatment after microbial infections, the response of
the immune system, PS cytotoxicity, but also the penetration depth of the light beam.
G. mellonella seems to be an appropriate model for the study of different aspects of aPDT
during infections caused by bacterial and fungal—mono- but also dual or multi-species
biofilms
[18,58,209212]
. Moreover, PDI can be tested in combination with other bioactive
molecules, including antimicrobial drugs [93,213].
Recently, scientists began to test the effectiveness of aPDT on G. mellonella infected
with C. albicans using different PSs, such as methylene blue [
214
], erythrosine, curcumin, or
toluidine blue [
121
,
209
,
213
]. In the dissertation of Dr. Dadi, the PS phloxine B was tested
Microorganisms 2023,11, 1455 6 of 21
for toxicity in Galleria larvae, and even a 0.5 mM concentration did not exhibit any effect on
G. mellonella survival [215].
The protocol for simple testing is as follows. After the inoculation of the larvae with
a cell suspension of a known density (this should usually be estimated in a preliminary
experiment for each microbial genus or species), the tested PS, diluted in sterile phosphate
buffer saline (PBS) to the desired concentration, is applied to the G.mellonella larvae, usually
by the inoculation method. Using a 10
µ
L Hamilton syringe, 10
µ
L aliquots of the cell
suspension are administrated into the hemocoel of each caterpillar via the proleg at the tail
end of the larva’s body, followed by the administration of the PS via the opposite proleg
(Figure 1) [
58
,
209
,
210
,
212
,
216
,
217
]. For some purposes, the PS can also be applied locally,
as described in a study by Figueiredo-Godoi et al. (2022) [
18
], who used G. mellonella for a
burn model infected with A. baumannii.
Microorganisms 2023, 11, x FOR PEER REVIEW 6 of 21
during infections caused by bacterial and fungalmono- but also dual or multi-species
biolms [18,58,209212]. Moreover, PDI can be tested in combination with other bioactive
molecules, including antimicrobial drugs [93,213].
Recently, scientists began to test the eectiveness of aPDT on G. mellonella infected
with C. albicans using dierent PSs, such as methylene blue [214], erythrosine, curcumin,
or toluidine blue [121,209,213]. In the dissertation of Dr. Dadi, the PS phloxine B was tested
for toxicity in Galleria larvae, and even a 0.5 mM concentration did not exhibit any eect
on G. mellonella survival [215].
The protocol for simple testing is as follows. After the inoculation of the larvae with
a cell suspension of a known density (this should usually be estimated in a preliminary
experiment for each microbial genus or species), the tested PS, diluted in sterile phosphate
buer saline (PBS) to the desired concentration, is applied to the G. mellonella larvae, usu-
ally by the inoculation method. Using a 10 µL Hamilton syringe, 10 µL aliquots of the cell
suspension are administrated into the hemocoel of each caterpillar via the proleg at the
tail end of the larva’s body, followed by the administration of the PS via the opposite pro-
leg (Figure 1) [58,209,210,212,216,217]. For some purposes, the PS can also be applied lo-
cally, as described in a study by Figueiredo-Godoi et al. (2022) [18], who used G. mellonella
for a burn model infected with A. baumannii.
Figure 1. Inoculation of G. mellonella larva with 1 mM methylene blue in PBS. (A) An injection station
for the simple manipulation of the larva during inoculation, which involves taping a lter paper
disc to the table and a 1000 µL-disposal tip onto the lter paper. (B) Injection of the larva: the G.
mellonella larva is gently held over the tip using the ngers, or tweezers, with the prolegs at the tail
end of the larva’s body visible. The needle is carefully inserted into a proleg, angling the needle
toward the head of the larva, and 10 µL of methylene blue in PBS is administrated. A dierent proleg
should be used for PS administration than for the previous inoculation of the pathogen to avoid
contamination. (C) During the release of the methylene blue, the larva visibly turns blue.
The application of the PS is followed by irradiation with light of an appropriate wave-
length, and the delivered energy is calculated, taking into account the duration of the ir-
radiation, to determine the total eectivity of irradiationuence. The PS application
should be approximately 1030 min before the irradiation, allowing the PS to penetrate
the tissue and nally the microorganisms. During irradiation, energy transfer from the PS
in the presence of oxygen results in the generation of ROS. One molecule of PS can activate
many atoms of activated oxygen. However, it should be considered that the diusion of
the activated oxygen is limited. Another limitation is the proximity to the PS, as objects
distant from it may be subjected to limited or no damage [209,213,218]. The irradiation of
G. mellonella is also a critical step, as it is important to ensure the proper delivery of the
light to cover the desired area of the insect body completely. For this purpose, the larva
should be maintained in a 24-well microtiter plate throughout the irradiation process
Figure 1.
Inoculation of G. mellonella larva with 1 mM methylene blue in PBS. (
A
) An injection station
for the simple manipulation of the larva during inoculation, which involves taping a filter paper disc
to the table and a 1000
µ
L-disposal tip onto the filter paper. (
B
) Injection of the larva: the G. mellonella
larva is gently held over the tip using the fingers, or tweezers, with the prolegs at the tail end of the
larva’s body visible. The needle is carefully inserted into a proleg, angling the needle toward the
head of the larva, and 10
µ
L of methylene blue in PBS is administrated. A different proleg should be
used for PS administration than for the previous inoculation of the pathogen to avoid contamination.
(C) During the release of the methylene blue, the larva visibly turns blue.
The application of the PS is followed by irradiation with light of an appropriate
wavelength, and the delivered energy is calculated, taking into account the duration of the
irradiation, to determine the total effectivity of irradiation—fluence. The PS application
should be approximately 10–30 min before the irradiation, allowing the PS to penetrate the
tissue and finally the microorganisms. During irradiation, energy transfer from the PS in
the presence of oxygen results in the generation of ROS. One molecule of PS can activate
many atoms of activated oxygen. However, it should be considered that the diffusion of
the activated oxygen is limited. Another limitation is the proximity to the PS, as objects
distant from it may be subjected to limited or no damage [
209
,
213
,
218
]. The irradiation
of G. mellonella is also a critical step, as it is important to ensure the proper delivery of
the light to cover the desired area of the insect body completely. For this purpose, the
larva should be maintained in a 24-well microtiter plate throughout the irradiation process
(Figure 2). To prevent the organism from moving around, it is advisable to perform the
irradiation of each larva separately, one by one, and to keep the larva inside its well using
forceps. After performing aPDT, the larvae are incubated in Petri dishes at the required
temperature (usually at 37
C in the dark). Every experiment must include a control group
of G. mellonella larvae that do not receive any injections to monitor the overall quality of the
Microorganisms 2023,11, 1455 7 of 21
larvae over the course of the experiment, as well as a PBS injection control group to ensure
that death was not due to trauma. The survival of aPDT-treated larvae is recorded daily
or hourly, according to a pathological scoring system proposed by Loh et al. (2013) [
78
]
taking into account a few attributes, such as movement activity, melanization, or cocoon
formation. Figure 2illustrates the irradiation of G. mellonella larvae with a red laser, and
how this is processed in the laboratory of Prof. Bujdákováet al.
Microorganisms 2023, 11, x FOR PEER REVIEW 7 of 21
(Figure 2). To prevent the organism from moving around, it is advisable to perform the
irradiation of each larva separately, one by one, and to keep the larva inside its well using
forceps. After performing aPDT, the larvae are incubated in Petri dishes at the required
temperature (usually at 37 °C in the dark). Every experiment must include a control group
of G. mellonella larvae that do not receive any injections to monitor the overall quality of
the larvae over the course of the experiment, as well as a PBS injection control group to
ensure that death was not due to trauma. The survival of aPDT-treated larvae is recorded
daily or hourly, according to a pathological scoring system proposed by Loh et al. (2013)
[78] taking into account a few aributes, such as movement activity, melanization, or co-
coon formation. Figure 2 illustrates the irradiation of G. mellonella larvae with a red laser,
and how this is processed in the laboratory of Prof. Bujdáková et al.
Figure 2. The process of the irradiation of G. mellonella larva with a red laser. (A) PDT assembly; (B)
the G. mellonella larva was positioned in the well of a 24-well microtiter plate; (C) irradiation of the
larva´s body with the red laser (λ = 660 nm, 190 mW/cm2). The distance between the larva and the
laser was 10 cm and the duration of irradiation was 2 min, which corresponded to an energy delivery
of 23 J.
The key factor in PDT (aPDI) eectiveness is PS, which must meet the compatibility
parameters and have high eciency. Absorption in the red and near-infrared spectrum is
also advantageous, as red light is relatively favorable to the treated host. The PS needs to
exhibit only local toxicity, even after light activation. A high level of ROS yield is also
assumed during irradiation [219221].
Phenothiazinium dyes are the most common PSs used in PDT performed on the G.
mellonella model [58,210,212,216,222]. During the administration of the desired PS into the
larva´s hemocoel, the body of the larva becomes colored, which is an accompanying
Figure 2.
The process of the irradiation of G. mellonella larva with a red laser. (
A
) PDT assembly;
(
B
) the G. mellonella larva was positioned in the well of a 24-well microtiter plate; (
C
) irradiation of
the larva´s body with the red laser (λ= 660 nm, 190 mW/cm2). The distance between the larva and
the laser was 10 cm and the duration of irradiation was 2 min, which corresponded to an energy
delivery of 23 J.
The key factor in PDT (aPDI) effectiveness is PS, which must meet the compatibility
parameters and have high efficiency. Absorption in the red and near-infrared spectrum
is also advantageous, as red light is relatively favorable to the treated host. The PS needs
to exhibit only local toxicity, even after light activation. A high level of ROS yield is also
assumed during irradiation [219221].
Phenothiazinium dyes are the most common PSs used in PDT performed on the
G. mellonella model [
58
,
210
,
212
,
216
,
222
]. During the administration of the desired PS into
the larva
´
s hemocoel, the body of the larva becomes colored, which is an accompanying
phenomenon. The intensity of the color depends on the concentration of the PS used. Over
the course of the experiment, the larvae excrete the dye and become discolored (Figure 3).
Microorganisms 2023,11, 1455 8 of 21
Microorganisms 2023, 11, x FOR PEER REVIEW 8 of 21
phenomenon. The intensity of the color depends on the concentration of the PS used. Over
the course of the experiment, the larvae excrete the dye and become discolored (Figure 3).
Figure 3. Testing the toxicity of 1 mM methylene blue on G. mellonella larvae. (A) Larvae without an
injectioncontrol group. (B) G. mellonella inoculated with 1 mM methylene blue in PBS. Immedi-
ately after inoculation, the larvae were visibly colored blue. (C) G. mellonella after 24 h. The larvae
slowly began to discolor, and their excrement was blue. (D) G. mellonella after 48 h. The discoloration
was progressing. (E) G. mellonella after 120 h. The larvae were completely discolored and resembled
the control group without any harmful eects.
G. mellonella larvae have been found to be versatile in several studies that evaluated
a PS used in aPDT [210,211,223]. De França et al. (2021) tested in vitro the anti-tumor eect
and skin permeation/retention of a green uorescence pyrene-based dye for aPDT, and
they used the G. mellonella model to determine PS toxicity [224]. Rigoo Caruso et al. (2021)
evaluated the antifungal activity of aPDT in vitro with dierent phenothiazinium PSs
(methylene blue, new methylene blue N, and new methylene blue N Zinc) in combination
with biosynthesized silver nanoparticles. The toxicity of all the tested compounds during
their study was veried in the G. mellonella model [225]. The tests performed in a study by
Malacarne et al. (2023) evaluated the toxicity of porphyrin PS on G. mellonella larvae and
its cytotoxicity on hemocytes. No dark toxicity of PS was observed, even at the highest
concentrations, and even with the longest incubation period (72 h). The intracellular lo-
calization of porphyrin PS was assessed using uorescence microscopy after the hemo-
cytes were isolated and collected from the hemolymph of inoculated larvae [226].
Nowadays, a relatively wide range of PSs are available, including phenothiazine
dyes, porphyrins, chlorines, and phthalocyanines. In addition to synthetic ones, natural
substances such as chlorophyllin, curcumin, and hypericin have also been studied
[18,209,217,227229].
The development of optimal light sources for PS is important for eective aPDT.
Many PSs used for in vivo testing are activated by a red light with a wavelength between
630 and 700 nm. The source of light is a light-emiing diode (LED light) or diode laser.
The irradiation itself must not aect the survival of the larvae [18,58,212,222,223,230,231].
During the interaction of the tissue with a light beam, most of the light is absorbed,
scaered, or transmied, and only 47% is reected. Pigmented tissue areas absorb light
preferentially compared to less pigmented ones [231]. aPDT can also be enhanced by
Figure 3.
Testing the toxicity of 1 mM methylene blue on G. mellonella larvae. (
A
) Larvae without an
injection—control group. (
B
)G. mellonella inoculated with 1 mM methylene blue in PBS. Immediately
after inoculation, the larvae were visibly colored blue. (
C
)G. mellonella after 24 h. The larvae slowly
began to discolor, and their excrement was blue. (
D
)G. mellonella after 48 h. The discoloration was
progressing. (
E
)G. mellonella after 120 h. The larvae were completely discolored and resembled the
control group without any harmful effects.
G. mellonella larvae have been found to be versatile in several studies that evaluated a
PS used in aPDT [
210
,
211
,
223
]. De França et al. (2021) tested
in vitro
the anti-tumor effect
and skin permeation/retention of a green fluorescence pyrene-based dye for aPDT, and
they used the G. mellonella model to determine PS toxicity [
224
]. Rigotto Caruso et al.
(2021) evaluated the antifungal activity of aPDT
in vitro
with different phenothiazinium PSs
(methylene blue, new methylene blue N, and new methylene blue N Zinc) in combination
with biosynthesized silver nanoparticles. The toxicity of all the tested compounds during
their study was verified in the G. mellonella model [
225
]. The tests performed in a study by
Malacarne et al. (2023) evaluated the toxicity of porphyrin PS on G. mellonella larvae and
its cytotoxicity on hemocytes. No dark toxicity of PS was observed, even at the highest
concentrations, and even with the longest incubation period (72 h). The intracellular local-
ization of porphyrin PS was assessed using fluorescence microscopy after the hemocytes
were isolated and collected from the hemolymph of inoculated larvae [226].
Nowadays, a relatively wide range of PSs are available, including phenothiazine dyes,
porphyrins, chlorines, and phthalocyanines. In addition to synthetic ones, natural substances
such as chlorophyllin, curcumin, and hypericin have also been studied [18,209,217,227229].
The development of optimal light sources for PS is important for effective aPDT. Many
PSs used for
in vivo
testing are activated by a red light with a wavelength between 630
and 700 nm. The source of light is a light-emitting diode (LED light) or diode laser. The
irradiation itself must not affect the survival of the larvae [18,58,212,222,223,230,231].
During the interaction of the tissue with a light beam, most of the light is absorbed,
scattered, or transmitted, and only 4–7% is reflected. Pigmented tissue areas absorb light
preferentially compared to less pigmented ones [
231
]. aPDT can also be enhanced by
increasing the PS concentration. However, higher concentrations of PS can result in the
formation of aggregates, leading to an optical shielding phenomenon that can reduce the
killing of microbial cells [232,233].
Microorganisms 2023,11, 1455 9 of 21
Merigo et al. (2017) studied the use of different laser energy densities (650 nm, 450 nm,
and 532 nm) with or without different types of PSs (toluidine blue, curcumin, and erythro-
sine) in C. albicans infections. The authors suggested that laser irradiation in combination
with an appropriate PS, and even the use of laser irradiation alone, were shown to be
effective at controlling candidiasis using the G. mellonella model [209].
In a study by Figueiredo-Godoi et al. (2019), red laser penetration, delivered at
different fluencies (660 nm, 6 and 15 J/cm
2
), and the distribution of light in the tissue of
G. mellonella larvae was investigated using a power meter and CCD camera. The images
were analyzed according to the interactive 3D Surface Plot plugin of the Image J program.
Subsequently, the concentration of the PS—methylene blue (100
µ
M) which allowed the
best light distribution over the thickness of the larvae’s body after administration was
chosen for the aPDT assays. The authors observed that without the PS, the beginning of
the light distribution in the cuticle occurred at 0.36 mm, and remained for up to 2.5mm. In
association with 100
µ
M methylene blue, the light distribution occurred at 0.27 mm and
extended up to 2.45 mm below the cuticle. These findings suggested that laser irradiation
in association with the proper PS can enhance light distribution in the cuticle [58].
Bispo et al. (2020) performed bacteria-targeted aPDT, which relied on the combination
of a bacteria-specific targeting agent and the light-induced generation of ROS by an ap-
propriate PS in G. mellonella. They conjugated the near-infrared PS IRDye700DX to a fully
human monoclonal antibody, specific to the immunodominant staphylococcal antigen A
(IsaA), creating a novel photo-immunoconjugate. They proved that aPDT with 1D9-700DX
was highly effective at treating G.mellonella infected with a methicillin-resistant strain.
Despite the observed relapse in the bacterial burden 48 h after aPDT, this relapse was not
lethal to the larvae, as there were increased survival rates (~80%) 72 h after treatment.
The authors suggested that the increased survival could be attributed to the innate larval
immune defenses. The authors concluded that aPDT with 1D9-700DX reduced the bacterial
burden to such an extent that the host’s immune responses could overcome infections
caused by multidrug-resistant S.aureus [234].
Chibebe et al. (2013) used G. mellonella for testing the effectiveness of aPDT in the
presence of methylene blue. They demonstrated the prolonged survival of G. mellonella after
infection with C. albicans. The fungal burden of G. mellonella hemolymph was reduced, and
the administration of fluconazole—either before or after exposing the larvae, infected with
fluconazole-resistant C. albicans, to aPDT—significantly prolonged their survival compared
to the control group. These findings suggested that aPDT combined with conventional
antimicrobial drugs could have a synergistic effect, representing an effective strategy for
the treatment of infections caused by resistant clinical strains [213].
The G. mellonella model has been used to identify the regulation of innate immunity
by aPDT [93,210,216,223]. Dos Santos et al. (2017) reported that aPDT activated the G. mel-
lonella immune system by increasing the circulation of hemocytes against Porphyromonas
gingivalis infection and by attenuating infection, prolonging the survival of the infected
group of larvae [
216
]. A study by Huang et al. (2020) [
223
] confirmed that aPDT had
immunomodulatory effects; they demonstrated that 5-aminolevulinic acid (ALA)-mediated
aPDT increased hemocyte density. Moreover, the extracted hemocytes after ALA-mediated
aPDT had increased susceptibility to C. albicans and S. aureus.
Paziani et al. (2019) found that the total hemocyte count after aPDT with phenoth-
iazinium PSs (methylene blue, new methylene blue, and pentacyclic phenothiazinium
photosensitizer S137) of infected G. mellonella increased in larvae hemolymph, whereas
the fungal burden was decreased. The increase in the cellular immune response was
correlated to the increase in larval survival and decrease in fungal burden. The survival
levels of infected larvae with Fusarium keratoplasticum were 70, 60, and 80% after aPDT with
methylene blue (1500
µ
M), new methylene blue (200
µ
M), and S137 (200
µ
M), respectively,
10 days after infection. The survival levels of larvae infected with Fusarium moniliforme
were 40, 10, and 100% after aPDT with methylene blue (1500
µ
M), new methylene blue
(200
µ
M), and S137 (200
µ
M), respectively, 10 days after infection. Thus, the larvae infected
Microorganisms 2023,11, 1455 10 of 21
with F. keratoplasticum and F. moniliforme, which were found to be resistant to itraconazole and
posaconazole, survived because the cellular immune system response of G. mellonella acted
effectively [210].
Table 1summarizes a list of published works studying the effectiveness of aPDT or PS
toxicity on G. mellonella using various conditions of aPDT, tested PSs, and microorganisms
selected for infection.
Table 1.
List of published works focused on testing PS toxicity or aPDT on microbial infection using
G. mellonella larvae as a model.
Photosensitizer Light Source Energy Microorganism Authors Reference
Methylene blue
0.2 mg/mL
660 nm red light device
composed of 48 LEDs 30 J/cm2Acinetobacter baumannii Figueiredo-Godoi
et al. (2022) [18]
Fotenticine
1.2 mg/mL
660 nm red light device
composed of 48 LEDs 30 J/cm2Acinetobacter baumannii Figueiredo-Godoi
et al. (2022) [18]
Methylene blue
75–600 µM660 nm red laser light 6 J/cm2and
15 J/cm2C. albicans Figueiredo-Godoi
et al. (2019) [58]
Methylene blue
1 mM
660 ±15 nm broadband
non-coherent red light
source
0.45–18 J/cm2Enterococcus faecium Chibebe Junior
et al. (2013) [93]
Erythrosine
100 µM
532 nm green diode laser
10 J/cm2C. albicans Merigo et al. (2017) [209]
Curcumin
100 µM
405 nm blue-violet diode
laser 10 J/cm2C. albicans Merigo et al. (2017) [209]
Toluidine blue
10 µM650 nm red diode laser 10 J/cm2C. albicans Merigo et al. (2017) [209]
Methylene blue
750–3000 µM
An array of
96 light-emitting diodes
with an emission peak at
635 nm and integrated
irradiance from 570 to
670 nm
15 J/cm2Fusarium keratoplasticum,
F. moniliforme
Paziani et al. (2019)
[210]
New methylene
blue N
100–400 µM
An array of
96 light-emitting diodes
with an emission peak at
635 nm and integrated
irradiance from 570 to
670 nm
15 J/cm2Fusarium keratoplasticum,
F. moniliforme
Paziani et al. (2019)
[210]
Pentacyclic
phenothiazinium
photosensitizer S137
100–400 µM
An array of
96 light-emitting diodes
with an emission peak at
635 nm and integrated
irradiance from 570 to
670 nm
15 J/cm2Fusarium keratoplasticum,
F. moniliforme
Paziani et al. (2019)
[210]
Curcumin
50 µg/mL 440–480 nm LED source 1.2 J/cm2Streptococcus mutants Sanches et al.
(2019) [211]
Diacetylcurcumin
50 µg/mL 440–480 nm LED source 1.2 J/cm2Streptococcus mutants Sanches et al.
(2019) [211]
Methylene blue
100 µM660 nm LED source 3–18 J/cm2Escherichia coli Garcez et al. (2020) [212]
Methylene blue
1 mM
660 ±15 nm broadband
non-coherent red light
source
0.45–18 J/cm2C. albicans Chibebe Junior
et al. (2013) [213]
Microorganisms 2023,11, 1455 11 of 21
Table 1. Cont.
Photosensitizer Light Source Energy Microorganism Authors Reference
Methylene blue
600 mM 660 nm red laser light 15 J/cm2Porphyromonas gingivalis Dos Santos et al.
(2017) [216]
Curcuma longa L.
Extract
100 mg/mL
Marques Meccatti
et al. (2022) [217]
Curcumin
200 µg/mL Marques Meccatti
et al. (2022) [217]
Methylene blue
Concentration not
specified
An array of
96 light-emitting diodes
with an emission peak at
635 nm
15 J/cm2C. albicans, C. auris Grizante Barião
et al. (2022) [222]
New methylene
blue N
Concentration not
specified
An array of
96 light-emitting diodes
with an emission peak at
635 nm
15 J/cm2C. albicans, C. auris Grizante Barião
et al. (2022) [222]
Toluidine blue O
Concentration not
specified
An array of
96 light-emitting diodes
with an emission peak at
635 nm
15 J/cm2C. albicans, C. auris Grizante Barião
et al. (2022) [222]
Pentacyclic
phenothiazinium
photosensitizer S137
Concentration not
specified
An array of
96 light-emitting diodes
with an emission peak at
635 nm
15 J/cm2C. albicans, C. auris Grizante Barião
et al. (2022) [222]
Methylene blue
10–500 mM
630 nm red
light-emitting diode
device
Not specified Fonsecaea monophora Huang et al. (2020) [223]
5-aminolevulinic acid
10–500 mM
630 nm red
light-emitting diode
device
Not specified Fonsecaea monophora Huang et al. (2020) [223]
4. Conclusions
The information summarized in this review points to the versatile use of G. mellonella
in biological research. This model has also been proven to be highly suitable for the study
of aPDT, despite some limitations, for example, the availability of oxygen in the tissues or
the delivery of light into the tissue, while achieving high efficiency in terms of irradiation.
Of course, the biocompatibility and photoactivity of the PS are the necessary conditions
for the overall effectiveness of aPDT. Many available and generally known techniques can
be adopted with G. mellonella in terms of the experiment design and expected results, but
the protocols must be optimized, taking into consideration the specificity of this model
organism. It is also necessary to think about the fact that the G. mellonella larvae must meet
the basic standard conditions for breeding and preservation to avoid discrepancies in the
obtained results. In summary, G. mellonella has great potential for experimental studies
of aPDT.
Author Contributions:
Conceptualization, H.B., L.B. and S.K.; writing—original draft preparation,
L.B. and S.K.; writing—review and editing, H.B., L.B. and S.K. All authors have read and agreed to
the published version of the manuscript.
Funding:
This review was supported by the EU’s Next Generation EU package through the Recovery
and Resilience Plan for Slovakia under project no. 09I03-03-V01-00105 from the Government Office of
the Slovak Republic, EU grant no. 952398—CEMBO, Call: H2020-WIDESPREAD-05-2020—Twinning,
Microorganisms 2023,11, 1455 12 of 21
the Slovak Research and Development Agency under contract APVV-21-0302, grant VEGA 2/0036/22
from the Ministry of Education, Science, Research, and Sport of the Slovak Republic.
Conflicts of Interest: The authors declare no conflict of interest.
References
1.
Lionakis, M.S. Drosophila and Galleria Insect Model Hosts: New Tools for the Study of Fungal Virulence, Pharmacology and
Immunology. Virulence 2011,2, 521–527. [CrossRef]
2.
Piatek, M.; Sheehan, G.; Kavanagh, K. Galleria mellonella: The Versatile Host for Drug Discovery, In Vivo Toxicity Testing and
Characterising Host-Pathogen Interactions. Antibiotics 2021,10, 1545. [CrossRef] [PubMed]
3.
Serrano, I.; Verdial, C.; Tavares, L.; Oliveira, M. The Virtuous Galleria mellonella Model for Scientific Experimentation. Antibiotics
2023,12, 505. [CrossRef] [PubMed]
4.
Asai, M.; Li, Y.; Newton, S.M.; Robertson, B.D.; Langford, P.R. Galleria mellonella—Intracellular Bacteria Pathogen Infection
Models: The Ins and Outs. FEMS Microbiol. Rev. 2023,47, fuad011. [CrossRef] [PubMed]
5.
Maurer, E.; Hörtnagl, C.; Lackner, M.; Grässle, D.; Naschberger, V.; Moser, P.; Segal, E.; Semis, M.; Lass-Flörl, C.; Binder, U. Galleria
mellonella as a Model System to Study Virulence Potential of Mucormycetes and Evaluation of Antifungal Treatment. Med. Mycol.
2019,57, 351–362. [CrossRef]
6.
Ménard, G.; Rouillon, A.; Cattoir, V.; Donnio, P.-Y. Galleria mellonella as a Suitable Model of Bacterial Infection: Past, Present and
Future. Front. Cell. Infect. Microbiol. 2021,11, 782733. [CrossRef]
7.
Banda, E. European Science Foundation Policy Briefing: Use of Animals in Research. Altern. Lab. Anim.
2000
,28, 743–749.
[CrossRef]
8.
EUR-Lex 32010L0063 EN EUR-Lex. Available online: https://eur-lex.europa.eu/legal-content/EN/TXT/?uri=celex%3A32010L0
063 (accessed on 17 April 2023).
9.
Hubrecht, R.C.; Carter, E. Carter The 3Rs and Humane Experimental Technique: Implementing Change. Animals
2019
,9, 754.
[CrossRef]
10.
Salinas, G.; Risi, G. Caenorhabditis elegans: Nature and Nurture Gift to Nematode Parasitologists. Parasitology
2018
,145, 979–987.
[CrossRef]
11.
Zhang, S.; Li, F.; Zhou, T.; Wang, G.; Li, Z. Caenorhabditis Elegans as a Useful Model for Studying Aging Mutations. Front.
Endocrinol. 2020,11, 554994. [CrossRef]
12. Tolwinski, N. Introduction: Drosophila—A Model System for Developmental Biology. JDB 2017,5, 9. [CrossRef]
13.
Staats, S.; Lüersen, K.; Wagner, A.E.; Rimbach, G. Drosophila melanogaster as a Versatile Model Organism in Food and Nutrition
Research. J. Agric. Food Chem. 2018,66, 3737–3753. [CrossRef] [PubMed]
14. Teame, T.; Zhang, Z.; Ran, C.; Zhang, H.; Yang, Y.; Ding, Q.; Xie, M.; Gao, C.; Ye, Y.; Duan, M.; et al. The Use of Zebrafish (Danio
rerio) as Biomedical Models. Anim. Front. 2019,9, 68–77. [CrossRef]
15.
Lim, S.; Kang, H.; Kwon, B.; Lee, J.P.; Lee, J.; Choi, K. Zebrafish (Danio rerio) as a Model Organism for Screening Nephrotoxic
Chemicals and Related Mechanisms. Ecotoxicol. Environ. Saf. 2022,242, 113842. [CrossRef]
16.
Allegra, E.; Titball, R.W.; Carter, J.; Champion, O.L. Galleria mellonella Larvae Allow the Discrimination of Toxic and Non-Toxic
Chemicals. Chemosphere 2018,198, 469–472. [CrossRef] [PubMed]
17.
Marcos-Zambrano, L.J.; Bordallo-Cardona, M.Á.; Borghi, E.; Falleni, M.; Tosi, D.; Muñoz, P.; Escribano, P.; Guinea, J. Candida
Isolates Causing Candidemia Show Different Degrees of Virulence in Galleria mellonella.Med. Mycol.
2020
,58, 83–92. [CrossRef]
18.
Figueiredo-Godoi, L.M.A.; Garcia, M.T.; Pinto, J.G.; Ferreira-Strixino, J.; Faustino, E.G.; Pedroso, L.L.C.; Junqueira, J.C. Antimicro-
bial Photodynamic Therapy Mediated by Fotenticine and Methylene Blue on Planktonic Growth, Biofilms, and Burn Infections of
Acinetobacter baumannii.Antibiotics 2022,11, 619. [CrossRef]
19.
Legéˇnová, K.; Kovalˇcíková, M.; ˇ
Cernáková, L.; Bujdáková, H. The Contribution of Photodynamic Inactivation vs. Corsodyl
Mouthwash to the Control of Streptococcus mutans Biofilms. Curr. Microbiol. 2020,77, 988–996. [CrossRef]
20.
ˇ
Cernáková, L.; Jordao, L.; Bujdáková, H. Impact of Farnesol and Corsodyl
®
on Candida Albicans Forming Dual Biofilm with
Streptococcus Mutans.Oral Dis. 2018,24, 1126–1131. [CrossRef]
21.
Dekkerová-Chupáˇcová, J.; Borghi, E.; Morace, G.; Bujdáková, H. Up-Regulation of Antimicrobial Peptides Gallerimycin and
Galiomicin in Galleria mellonella Infected with Candida Yeasts Displaying Different Virulence Traits. Mycopathologia
2018
,183,
935–940. [CrossRef]
22.
Singkum, P.; Suwanmanee, S.; Pumeesat, P.; Luplertlop, N. A Powerful in Vivo Alternative Model in Scientific Research: Galleria
mellonella.AMicr 2019,66, 31–55. [CrossRef] [PubMed]
23.
Asai, M.; Sheehan, G.; Li, Y.; Robertson, B.D.; Kavanagh, K.; Langford, P.R.; Newton, S.M. Innate Immune Responses of Galleria
mellonella to Mycobacterium bovis BCG Challenge Identified Using Proteomic and Molecular Approaches. Front. Cell. Infect.
Microbiol. 2021,11, 619981. [CrossRef]
24.
Wojda, I.; Staniec, B.; Sułek, M.; Kordaczuk, J. The Greater Wax Moth Galleria mellonella: Biology and Use in Immune Studies.
Pathog. Dis. 2020,78, ftaa057. [CrossRef] [PubMed]
Microorganisms 2023,11, 1455 13 of 21
25.
Chupáˇcová, J.; Borghi, E.; Morace, G.; Los, A.; Bujdáková, H. Anti-Biofilm Activity of Antibody Directed against Surface Antigen
Complement Receptor 3-Related Protein—Comparison of Candida albicans and Candida dubliniensis.Pathog. Dis.
2018
,76, ftx127.
[CrossRef] [PubMed]
26.
Wainwright, M.; Maisch, T.; Nonell, S.; Plaetzer, K.; Almeida, A.; Tegos, G.P.; Hamblin, M.R. Photoantimicrobials—Are We Afraid
of the Light? Lancet Infect. Dis. 2017,17, e49–e55. [CrossRef]
27.
Abrahamse, H.; Hamblin, M.R. New Photosensitizers for Photodynamic Therapy. Biochem. J.
2016
,473, 347–364. [CrossRef]
[PubMed]
28.
Cieplik, F.; Deng, D.; Crielaard, W.; Buchalla, W.; Hellwig, E.; Al-Ahmad, A.; Maisch, T. Antimicrobial Photodynamic Therapy—
What We Know and What We Don’t. Crit. Rev. Microbiol. 2018,44, 571–589. [CrossRef]
29.
Firacative, C.; Khan, A.; Duan, S.; Ferreira-Paim, K.; Leemon, D.; Meyer, W. Rearing and Maintenance of Galleria mellonella and Its
Application to Study Fungal Virulence. JoF 2020,6, 130. [CrossRef]
30.
Smith, T.L. External Morphology of the Larva, Pupa, and Adult of the Wax Moth, Galleria mellonella L. J. Kans. Entomol. Soc.
2023
,
38, 287–310.
31. Swamy, B.C.H. Bionomics and Biometrics of Greater Wax Moth Galleria mellonella Linnaeus. Asian J. Bio Sci. 2007,3, 49–51.
32. Ellis, J.D.; Graham, J.R.; Mortensen, A. Standard Methods for Wax Moth Research. J. Apic. Res. 2013,52, 1–17. [CrossRef]
33.
Hosamani, V.; Hanumantha Swamy, B.C.; Kattimani, K.N.; Kalibavi, C.M. Studies on Biology of Greater Wax Moth (Galleria
mellonella L.). Int. J. Curr. Microbiol. App. Sci. 2017,6, 3811–3815. [CrossRef]
34.
Desai, A.V.; Siddhapara, M.R.; Patel, P.K.; Prajapati, A.P. Biology of Greater Wax Moth, Galleria mellonella L. on Artificial Diet.
J. Exp. Zool. India 2019,22, 1267–1272.
35.
Brennan, M.; Thomas, D.Y.; Whiteway, M.; Kavanagh, K. Correlation between Virulence of Candida albicans Mutants in Mice and
Galleria mellonella Larvae. FEMS Immunol. Med. Microbiol. 2002,34, 153–157. [CrossRef] [PubMed]
36.
Fuchs, B.B.; Mylonakis, E. Using Non-Mammalian Hosts to Study Fungal Virulence and Host Defense. Curr. Opin. Microbiol.
2006,9, 346–351. [CrossRef] [PubMed]
37.
Iqbal, H.; Wright, C.L.; Jones, S.; da Silva, G.R.; McKillen, J.; Gilmore, B.F.; Kavanagh, O.; Green, B.D. Extracts of Sida Cordifolia
Contain Polysaccharides Possessing Immunomodulatory Activity and Rosmarinic Acid Compounds with Antibacterial Activity.
BMC Complement Med. Ther. 2022,22, 27. [CrossRef]
38.
Durieux, M.-F.; Melloul, É.; Jemel, S.; Roisin, L.; Dardé, M.-L.; Guillot, J.; Dannaoui, É.; Botterel, F. Galleria mellonella as a Screening
Tool to Study Virulence Factors of Aspergillus fumigatus.Virulence 2021,12, 818–834. [CrossRef]
39.
Tao, Y.; Duma, L.; Rossez, Y. Galleria mellonella as a Good Model to Study Acinetobacter baumannii Pathogenesis. Pathogens
2021
,10,
1483. [CrossRef]
40.
Malmquist, J.A.; Rogan, M.R.; McGillivray, S.M. Galleria mellonella as an Infection Model for Bacillus anthracis Sterne. Front. Cell
Infect Microbiol. 2019,9, 360. [CrossRef]
41.
Firacative, C.; Duan, S.; Meyer, W. Galleria mellonella Model Identifies Highly Virulent Strains among All Major Molecular Types
of Cryptococcus gattii.PLoS ONE 2014,9, e105076. [CrossRef]
42.
Borman, A.M. Of Mice and Men and Larvae: Galleria mellonella to Model the Early Host-Pathogen Interactions after Fungal
Infection. Virulence 2018,9, 9–12. [CrossRef]
43.
Mowlds, P.; Kavanagh, K. Effect of Pre-Incubation Temperature on Susceptibility of Galleria mellonella Larvae to Infection by
Candida albicans.Mycopathologia 2008,165, 5–12. [CrossRef] [PubMed]
44.
Joyce, S.A.; Gahan, C.G.M. Molecular Pathogenesis of Listeria Monocytogenes in the Alternative Model Host Galleria mellonella.
Microbiology 2010,156, 3456–3468. [CrossRef] [PubMed]
45.
Kloezen, W.; van Helvert-van Poppel, M.; Fahal, A.H.; van de Sande, W.W. A Madurella mycetomatis Grain Model in Galleria
mellonella Larvae. PLOS Negl. Trop. Dis. 2015,9, e0003926. [CrossRef] [PubMed]
46.
Vogel, H.; Altincicek, B.; Glöckner, G.; Vilcinskas, A. A Comprehensive Transcriptome and Immune-Gene Repertoire of the
Lepidopteran Model Host Galleria mellonella.BMC Genom. 2011,12, 308. [CrossRef]
47.
Lange, A.; Schäfer, A.; Bender, A.; Steimle, A.; Beier, S.; Parusel, R.; Frick, J.-S. Galleria mellonella: A Novel Invertebrate Model to
Distinguish Intestinal Symbionts from Pathobionts. Front. Immunol. 2018,9, 2114. [CrossRef]
48.
Lange, A.; Beier, S.; Huson, D.H.; Parusel, R.; Iglauer, F.; Frick, J.-S. Genome Sequence of Galleria mellonella (Greater Wax Moth).
Genome Announc. 2018,6, e01220-17. [CrossRef]
49.
Cunha, F.; Burne, A.; Casaro, S.; Brown, M.B.; Bisinotto, R.S.; Galvao, K.N. Establishing Galleria mellonella as an Invertebrate
Model for the Emerging Multi-Host Pathogen Helcococcus Ovis.Virulence 2023,14, 2186377. [CrossRef]
50.
Ahlawat, S.; Sharma, K.K. Lepidopteran Insects: Emerging Model Organisms to Study Infection by Enteropathogens. Folia
Microbiol. 2023,68, 181–196. [CrossRef]
51.
Yang, Z.; Zhang, F.; Li, D.; Wang, S.; Pang, Z.; Chen, L.; Li, R.; Shi, D. Correlation Between Drug Resistance and Virulence of
Candida Isolates from Patients with Candidiasis. IDR 2022,15, 7459–7473. [CrossRef]
52.
Fuchs, B.B.; O’Brien, E.; El Khoury, J.B.; Mylonakis, E. Methods for Using Galleria mellonella as a Model Host to Study Fungal
Pathogenesis. Virulence 2010,1, 475–482. [CrossRef] [PubMed]
53.
Hesketh-Best, P.J.; Mouritzen, M.V.; Shandley-Edwards, K.; Billington, R.A.; Upton, M. Galleria mellonella Larvae Exhibit a
Weight-Dependent Lethal Median Dose When Infected with Methicillin-Resistant Staphylococcus aureus.Pathog. Dis.
2021
,79,
ftab003. [CrossRef]
Microorganisms 2023,11, 1455 14 of 21
54.
Altunta¸s, H.; Gwokyalya, R.; Bayram, N. Immunotoxic Effects of Force-Fed Ethephon on Model Organism Galleria mellonella
(Lepidoptera: Pyralidae). Drug Chem. Toxicol. 2022,45, 1761–1768. [CrossRef]
55.
Jemel, S.; Guillot, J.; Kallel, K.; Botterel, F.; Dannaoui, E. Galleria mellonella for the Evaluation of Antifungal Efficacy against
Medically Important Fungi, a Narrative Review. Microorganisms 2020,8, 390. [CrossRef] [PubMed]
56.
Jin, X.; Zhang, M.; Lu, J.; Duan, X.; Chen, J.; Liu, Y.; Chang, W.; Lou, H. Hinokitiol Chelates Intracellular Iron to Retard Fungal
Growth by Disturbing Mitochondrial Respiration. J. Adv. Res. 2021,34, 65–77. [CrossRef]
57.
Stempinski, P.; Smith, D.; Casadevall, A. Cryptococcus neoformans Virulence Assay Using a Galleria mellonella Larvae Model System.
Bio. Protoc. 2022,12, e4480. [CrossRef] [PubMed]
58.
Figueiredo-Godoi, L.M.A.; Menezes, R.T.; Carvalho, J.S.; Garcia, M.T.; Segundo, A.G.; Jorge, A.O.C.; Junqueira, J.C. Exploring the
Galleria mellonella Model to Study Antifungal Photodynamic Therapy. Photodiagnosis Photodyn. Ther.
2019
,27, 66–73. [CrossRef]
[PubMed]
59.
Güntzel, P.; Nagel, C.; Weigelt, J.; Betts, J.W.; Pattrick, C.A.; Southam, H.M.; La Ragione, R.M.; Poole, R.K.; Schatzschneider, U.
Biological Activity of Manganese(I) Tricarbonyl Complexes on Multidrug-Resistant Gram-Negative Bacteria: From Functional
Studies to in Vivo Activity in Galleria mellonella.Metallomics 2019,11, 2033–2042. [CrossRef]
60.
Thomaz, L.; Gustavo De Almeida, L.; Silva, F.R.O.; Cortez, M.; Taborda, C.P.; Spira, B. In Vivo Activity of Silver Nanoparticles
Against Pseudomonas aeruginosa Infection in Galleria mellonella.Front. Microbiol. 2020,11, 582107. [CrossRef]
61.
McCloskey, A.P.; Lee, M.; Megaw, J.; McEvoy, J.; Coulter, S.M.; Pentlavalli, S.; Laverty, G. Investigating the In Vivo Antimicrobial
Activity of a Self-Assembling Peptide Hydrogel Using a Galleria mellonella Infection Model. ACS Omega
2019
,4, 2584–2589.
[CrossRef]
62.
Mannix-Fisher, E.; McLean, S. The Antimicrobial Activity of Silver Acetate against Acinetobacter baumannii in a Galleria mellonella
Infection Model. PeerJ 2021,9, e11196. [CrossRef] [PubMed]
63.
Scalfaro, C.; Iacobino, A.; Nardis, C.; Franciosa, G. Galleria mellonella as an in Vivo Model for Assessing the Protective Activity of
Probiotics against Gastrointestinal Bacterial Pathogens. FEMS Microbiol. Lett. 2017,364, fnx064. [CrossRef]
64.
Moman, R.; O’Neill, C.A.; Ledder, R.G.; Cheesapcharoen, T.; McBain, A.J. Mitigation of the Toxic Effects of Periodontal Pathogens
by Candidate Probiotics in Oral Keratinocytes, and in an Invertebrate Model. Front. Microbiol. 2020,11, 999. [CrossRef]
65.
Li, M.; Carpenter, C.E.; Broadbent, J.R. Organic Acid Exposure Enhances Virulence in Some Listeria Monocytogenes Strains Using
the Galleria mellonella Infection Model. Front. Microbiol. 2021,12, 675241. [CrossRef] [PubMed]
66.
Mukherjee, K.; Hain, T.; Fischer, R.; Chakraborty, T.; Vilcinskas, A. Brain Infection and Activation of Neuronal Repair Mechanisms
by the Human Pathogen Listeria monocytogenes in the Lepidopteran Model Host Galleria mellonella.Virulence
2013
,4, 324–332.
[CrossRef] [PubMed]
67.
Mariotti, M.; Lombardini, G.; Rizzo, S.; Scarafile, D.; Modesto, M.; Truzzi, E.; Benvenuti, S.; Elmi, A.; Bertocchi, M.; Fiorentini, L.;
et al. Potential Applications of Essential Oils for Environmental Sanitization and Antimicrobial Treatment of Intensive Livestock
Infections. Microorganisms 2022,10, 822. [CrossRef]
68.
Kay, S.; Edwards, J.; Brown, J.; Dixon, R. Galleria mellonella Infection Model Identifies Both High and Low Lethality of Clostridium
Perfringens Toxigenic Strains and Their Response to Antimicrobials. Front. Microbiol. 2019,10, 1281. [CrossRef] [PubMed]
69.
Benthall, G.; Touzel, R.E.; Hind, C.K.; Titball, R.W.; Sutton, J.M.; Thomas, R.J.; Wand, M.E. Evaluation of Antibiotic Efficacyagainst
Infections Caused by Planktonic or Biofilm Cultures of Pseudomonas aeruginosa and Klebsiella kneumoniae in Galleria mellonella.Int.
J. Antimicrob. Agents 2015,46, 538–545. [CrossRef]
70.
Aparecida Procópio Gomes, L.; Alves Figueiredo, L.M.; Luiza do Rosário Palma, A.; Corrêa Geraldo, B.M.; Isler Castro, K.C.;
Ruano de Oliveira Fugisaki, L.; Jorge, A.O.C.; de Oliveira, L.D.; Junqueira, J.C. Punica granatum L. (Pomegranate) Extract: In Vivo
Study of Antimicrobial Activity against Porphyromonas gingivalis in Galleria mellonella Model. Sci. World J.
2016
,2016, 8626987.
[CrossRef]
71.
Lázár, V.; Snitser, O.; Barkan, D.; Kishony, R. Antibiotic Combinations Reduce Staphylococcus aureus Clearance. Nature
2022
,610,
540–546. [CrossRef]
72.
Peleg, A.Y.; Monga, D.; Pillai, S.; Mylonakis, E.; Moellering, R.C.; Eliopoulos, G.M. Reduced Susceptibility to Vancomycin
Influences Pathogenicity in Staphylococcus aureus Infection. J. Infect Dis. 2009,199, 532–536. [CrossRef] [PubMed]
73.
Quiblier, C.; Seidl, K.; Roschitzki, B.; Zinkernagel, A.S.; Berger-Bächi, B.; Senn, M.M. Secretome Analysis Defines the Major Role
of SecDF in Staphylococcus aureus Virulence. PLoS ONE 2013,8, e63513. [CrossRef] [PubMed]
74.
Desbois, A.P.; Coote, P.J. Wax Moth Larva (Galleria mellonella): An in Vivo Model for Assessing the Efficacy of Antistaphylococcal
Agents. J. Antimicrob. Chemother. 2011,66, 1785–1790. [CrossRef] [PubMed]
75.
Materazzi, A.; Bottai, D.; Campobasso, C.; Klatt, A.-B.; Cesta, N.; De Masi, M.; Trampuz, A.; Tavanti, A.; Di Luca, M. Phage-Based
Control of Methicillin Resistant Staphylococcus aureus in a Galleria mellonella Model of Implant-Associated Infection. Int. J. Mol. Sci.
2022,23, 14514. [CrossRef] [PubMed]
76.
Sheehan, G.; Dixon, A.; Kavanagh, K. Utilization of Galleria mellonella Larvae to Characterize the Development of Staphylococcus
aureus Infection. Microbiology 2019,165, 863–875. [CrossRef]
77.
Mishra, B.; Khader, R.; Felix, L.O.; Frate, M.; Mylonakis, E.; Meschwitz, S.; Fuchs, B.B. A Substituted Diphenyl Amide Based
Novel Scaffold Inhibits Staphylococcus aureus Virulence in a Galleria mellonella Infection Model. Front. Microbiol.
2021
,12, 723133.
[CrossRef]
Microorganisms 2023,11, 1455 15 of 21
78.
Loh, J.M.S.; Adenwalla, N.; Wiles, S.; Proft, T. Galleria mellonella Larvae as an Infection Model for Group A Streptococcus.Virulence
2013,4, 419–428. [CrossRef]
79.
Olsen, R.J.; Watkins, M.E.; Cantu, C.C.; Beres, S.B.; Musser, J.M. Virulence of Serotype M3 Group A Streptococcus Strains in Wax
Worms (Galleria mellonella Larvae). Virulence 2011,2, 111–119. [CrossRef]
80.
Tsai, C.J.-Y.; Loh, J.M.S.; Proft, T. The Use of Galleria mellonella (Wax Moth) as an Infection Model for Group A Streptococcus.
Methods Mol. Biol. 2020,2136, 279–286. [CrossRef]
81.
Cools, F.; Torfs, E.; Aizawa, J.; Vanhoutte, B.; Maes, L.; Caljon, G.; Delputte, P.; Cappoen, D.; Cos, P. Optimization and
Characterization of a Galleria mellonella Larval Infection Model for Virulence Studies and the Evaluation of Therapeutics Against
Streptococcus pneumoniae.Front. Microbiol. 2019,10, 311. [CrossRef]
82.
Evans, B.A.; Rozen, D.E. A Streptococcus pneumoniae Infection Model in Larvae of the Wax Moth Galleria mellonella.Eur. J. Clin.
Microbiol. Infect Dis. 2012,31, 2653–2660. [CrossRef] [PubMed]
83.
Leal, J.T.; Primon-Barros, M.; de Carvalho Robaina, A.; Pizzutti, K.; Mott, M.P.; Trentin, D.S.; Dias, C.A.G. Streptococcus pneumoniae
Serotype 19A from Carriers and Invasive Disease: Virulence Gene Profile and Pathogenicity in a Galleria mellonella Model. Eur. J.
Clin. Microbiol. Infect Dis. 2023,42, 399–411. [CrossRef] [PubMed]
84.
Dos Santos, J.D.; de Oliveira Fugisaki, L.R.; Medina, R.P.; Scorzoni, L.; de SáAlves, M.; de Barros, P.P.; Ribeiro, F.C.; Fuchs, B.B.;
Mylonakis, E.; Silva, D.H.S.; et al. Streptococcus mutans Secreted Products Inhibit Candida albicans Induced Oral Candidiasis. Front.
Microbiol. 2020,11, 1605. [CrossRef]
85.
Buckley, A.A.; Faustoferri, R.C.; Quivey, R.G.
β
-Phosphoglucomutase Contributes to Aciduricity in Streptococcus mutans.Microbi-
ology 2014,160, 818–827. [CrossRef]
86.
Andresen, S.; de Mojana di Cologna, N.; Archer-Hartmann, S.; Rogers, A.M.; Samaddar, S.; Ganguly, T.; Black, I.M.; Glushka, J.;
Ng, K.K.S.; Azadi, P.; et al. Involvement of the Streptococcus mutans PgfE and GalE 4-Epimerases in Protein Glycosylation, Carbon
Metabolism, and Cell Division. Glycobiology 2023,33, 245–259. [CrossRef]
87.
Mukherjee, K.; Mraheil, M.A.; Silva, S.; Müller, D.; Cemic, F.; Hemberger, J.; Hain, T.; Vilcinskas, A.; Chakraborty, T. Anti-Listeria
Activities of Galleria mellonella Hemolymph Proteins. Appl. Environ. Microbiol. 2011,77, 4237–4240. [CrossRef]
88.
Mukherjee, K.; Altincicek, B.; Hain, T.; Domann, E.; Vilcinskas, A.; Chakraborty, T. Galleria mellonella as a Model System for
Studying Listeria Pathogenesis. Appl. Environ. Microbiol. 2010,76, 310–317. [CrossRef] [PubMed]
89.
La Rosa, S.L.; Casey, P.G.; Hill, C.; Diep, D.B.; Nes, I.F.; Brede, D.A. In Vivo Assessment of Growth and Virulence Gene Expression
during Commensal and Pathogenic Lifestyles of LuxABCDE-Tagged Enterococcus faecalis Strains in Murine Gastrointestinal and
Intravenous Infection Models. Appl. Environ. Microbiol. 2013,79, 3986–3997. [CrossRef]
90.
La Rosa, S.L.; Diep, D.B.; Nes, I.F.; Brede, D.A. Construction and Application of a LuxABCDE Reporter System for Real-Time
Monitoring of Enterococcus faecalis Gene Expression and Growth. Appl. Environ. Microbiol. 2012,78, 7003–7011. [CrossRef]
91.
Lam, L.N.; Brunson, D.N.; Kajfasz, J.K.; Lemos, J.A. Methods for Using the Galleria mellonella Invertebrate Model to Probe
Enterococcus faecalis Pathogenicity. Methods Mol. Biol. 2022,2427, 177–183. [CrossRef] [PubMed]
92.
Thieme, L.; Hartung, A.; Makarewicz, O.; Pletz, M.W. In Vivo Synergism of Ampicillin, Gentamicin, Ceftaroline and Ceftriaxone
against Enterococcus Faecalis Assessed in the Galleria mellonella Infection Model. J. Antimicrob. Chemother.
2020
,75, 2173–2181.
[CrossRef]
93.
Chibebe Junior, J.; Fuchs, B.B.; Sabino, C.P.; Junqueira, J.C.; Jorge, A.O.C.; Ribeiro, M.S.; Gilmore, M.S.; Rice, L.B.; Tegos, G.P.;
Hamblin, M.R.; et al. Photodynamic and Antibiotic Therapy Impair the Pathogenesis of Enterococcus faecium in a Whole Animal
Insect Model. PLoS ONE 2013,8, e55926. [CrossRef] [PubMed]
94.
Lebreton, F.; Le Bras, F.; Reffuveille, F.; Ladjouzi, R.; Giard, J.-C.; Leclercq, R.; Cattoir, V. Galleria mellonella as a Model for Studying
Enterococcus faecium Host Persistence. J. Mol. Microbiol. Biotechnol. 2011,21, 191–196. [CrossRef] [PubMed]
95.
Pradal, I.; Casado, A.; Del Rio, B.; Rodriguez-Lucas, C.; Fernandez, M.; Alvarez, M.A.; Ladero, V. Enterococcus Faecium
Bacteriophage VB_EfaH_163, a New Member of the Herelleviridae Family, Reduces the Mortality Associated with an E. Faecium
VanR Clinical Isolate in a Galleria mellonella Animal Model. Viruses 2023,15, 179. [CrossRef]
96.
Lagatolla, C.; Mehat, J.W.; La Ragione, R.M.; Luzzati, R.; Di Bella, S. In Vitro and In Vivo Studies of Oritavancin and Fosfomycin
Synergism against Vancomycin-Resistant Enterococcus faecium.Antibiotics 2022,11, 1334. [CrossRef] [PubMed]
97.
Meir, M.; Grosfeld, T.; Barkan, D. Establishment and Validation of Galleria mellonella as a Novel Model Organism to Study
Mycobacterium Abscessus Infection, Pathogenesis, and Treatment. Antimicrob. Agents Chemother.
2018
,62, e02539-17. [CrossRef]
98.
Meir, M.; Bifani, P.; Barkan, D. The Addition of Avibactam Renders Piperacillin an Effective Treatment for Mycobacterium
Abscessus Infection in an in Vivo Model. Antimicrob. Resist Infect Control 2018,7, 151. [CrossRef]
99.
Asai, M.; Li, Y.; Spiropoulos, J.; Cooley, W.; Everest, D.J.; Kendall, S.L.; Martín, C.; Robertson, B.D.; Langford, P.R.; Newton,
S.M. Galleria mellonella as an Infection Model for the Virulent Mycobacterium tuberculosis H37Rv. Virulence
2022
,13, 1543–1557.
[CrossRef]
100.
Li, Y.; Spiropoulos, J.; Cooley, W.; Khara, J.S.; Gladstone, C.A.; Asai, M.; Bossé, J.T.; Robertson, B.D.; Newton, S.M.; Langford, P.R.
Galleria mellonella—A Novel Infection Model for the Mycobacterium tuberculosis Complex. Virulence
2018
,9, 1126–1137. [CrossRef]
101.
Asai, M.; Li, Y.; Khara, J.S.; Gladstone, C.A.; Robertson, B.D.; Langford, P.R.; Newton, S.M. Use of the Invertebrate Galleria
mellonella as an Infection Model to Study the Mycobacterium tuberculosis Complex. J. Vis. Exp. 2019,148, e59703. [CrossRef]
102.
Briffotaux, J.; Xu, Y.; Huang, W.; Hui, Z.; Wang, X.; Gicquel, B.; Liu, S. A Hydrazine-Hydrazone Adamantine Compound Shows
Antimycobacterial Activity and Is a Probable Inhibitor of MmpL3. Molecules 2022,27, 7130. [CrossRef]
Microorganisms 2023,11, 1455 16 of 21
103.
Chen, R.Y.; Keddie, B.A. The Galleria mellonella-Enteropathogenic Escherichia coli Model System: Characterization of Pathogen
Virulence and Insect Immune Responses. J. Insect. Sci. 2021,21, 7. [CrossRef] [PubMed]
104.
Prasastha Ram, V.; Yasur, J.; Abishad, P.; Unni, V.; Purushottam Gourkhede, D.; Nishanth, M.A.D.; Niveditha, P.; Vergis, J.; Singh
Malik, S.V.; Kullaiah, B.; et al. Antimicrobial Efficacy of Green Synthesized Nanosilver with Entrapped Cinnamaldehyde against
Multi-Drug-Resistant Enteroaggregative Escherichia coli in Galleria mellonella.Pharmaceutics 2022,14, 1924. [CrossRef] [PubMed]
105.
Guerrieri, C.G.; Pereira, M.F.; Galdino, A.C.M.; Dos Santos, A.L.S.; Elias, W.P.; Schuenck, R.P.; Spano, L.C. Typical and Atypical
Enteroaggregative Escherichia coli Are Both Virulent in the Galleria mellonella Model. Front. Microbiol.
2019
,10, 1791. [CrossRef]
[PubMed]
106.
Williamson, D.A.; Mills, G.; Johnson, J.R.; Porter, S.; Wiles, S. In Vivo Correlates of Molecularly Inferred Virulence among
Extraintestinal Pathogenic Escherichia coli (ExPEC) in the Wax Moth Galleria mellonella Model System. Virulence
2014
,5, 388–393.
[CrossRef]
107.
Viegas, S.C.; Mil-Homens, D.; Fialho, A.M.; Arraiano, C.M. The Virulence of Salmonella enterica Serovar Typhimurium in the
Insect Model Galleria mellonella Is Impaired by Mutations in RNase E and RNase III. Appl. Environ. Microbiol.
2013
,79, 6124–6133.
[CrossRef]
108.
Kosznik-Kwa´snicka, K.; Stasiłoj´c, M.; Grabowski, Ł.; Zdrojewska, K.; egrzyn, G.; egrzyn, A. Efficacy and Safety of Phage
Therapy against Salmonella enterica Serovars Typhimurium and Enteritidis Estimated by Using a Battery of in Vitro Tests and the
Galleria mellonella Animal Model. Microbiol. Res. 2022,261, 127052. [CrossRef]
109.
Figueiredo, R.; Card, R.; Nunes, C.; AbuOun, M.; Bagnall, M.C.; Nunez, J.; Mendonça, N.; Anjum, M.F.; da Silva, G.J. Virulence
Characterization of Salmonella enterica by a New Microarray: Detection and Evaluation of the Cytolethal Distending Toxin Gene
Activity in the Unusual Host S. Typhimurium. PLoS ONE 2015,10, e0135010. [CrossRef]
110.
Insua, J.L.; Llobet, E.; Moranta, D.; Pérez-Gutiérrez, C.; Tomás, A.; Garmendia, J.; Bengoechea, J.A. Modeling Klebsiella kneumoniae
Pathogenesis by Infection of the Wax Moth Galleria mellonella.Infect. Immun. 2013,81, 3552–3565. [CrossRef]
111.
Sugeçti, S. Pathophysiological Effects of Klebsiella kneumoniae Infection on Galleria mellonella as an Invertebrate Model Organism.
Arch. Microbiol. 2021,203, 3509–3517. [CrossRef]
112.
Russo, T.A.; MacDonald, U. The Galleria mellonella Infection Model Does Not Accurately Differentiate between Hypervirulent and
Classical Klebsiella kneumoniae.mSphere 2020,5, e00850-19. [CrossRef]
113.
Pu, M.; Han, P.; Zhang, G.; Liu, Y.; Li, Y.; Li, F.; Li, M.; An, X.; Song, L.; Chen, Y.; et al. Characterization and Comparative
Genomics Analysis of a New Bacteriophage BUCT610 against Klebsiella kneumoniae and Efficacy Assessment in Galleria mellonella
Larvae. Int. J. Mol. Sci. 2022,23, 8040. [CrossRef]
114.
Khalil, M.A.F.; Ahmed, F.A.; Elkhateeb, A.F.; Mahmoud, E.E.; Ahmed, M.I.; Ahmed, R.I.; Hosni, A.; Alghamdi, S.; Kabrah, A.;
Dablool, A.S.; et al. Virulence Characteristics of Biofilm-Forming Acinetobacter baumannii in Clinical Isolates Using a Galleria
mellonella Model. Microorganisms 2021,9, 2365. [CrossRef]
115.
Park, J.; Kim, M.; Shin, B.; Kang, M.; Yang, J.; Lee, T.K.; Park, W. A Novel Decoy Strategy for Polymyxin Resistance in Acinetobacter
baumannii.Elife 2021,10, e66988. [CrossRef]
116.
Yang, H.; Chen, G.; Hu, L.; Liu, Y.; Cheng, J.; Li, H.; Ye, Y.; Li, J. In Vivo Activity of Daptomycin/Colistin Combination Therapy in
aGalleria mellonella Model of Acinetobacter baumannii Infection. Int. J. Antimicrob. Agents 2015,45, 188–191. [CrossRef]
117.
Miller, S.; Goy, K.; She, R.; Spellberg, B.; Luna, B. Antimicrobial Susceptibility Testing Performed in RPMI 1640 Reveals
Azithromycin Efficacy against Carbapenem-Resistant Acinetobacter baumannii and Predicts In Vivo Outcomes in Galleria mellonella.
Antimicrob. Agents Chemother. 2023,67, e0132022. [CrossRef] [PubMed]
118.
Thelaus, J.; Lundmark, E.; Lindgren, P.; Sjödin, A.; Forsman, M. Galleria mellonella Reveals Niche Differences Between Highly
Pathogenic and Closely Related Strains of Francisella Spp. Front. Cell Infect. Microbiol. 2018,8, 188. [CrossRef] [PubMed]
119.
Brodmann, M.; Schnider, S.T.; Basler, M. Type VI Secretion System and Its Effectors PdpC, PdpD, and OpiA Contribute to
Francisella Virulence in Galleria mellonella Larvae. Infect. Immun. 2021,89, e0057920. [CrossRef] [PubMed]
120.
Propst, C.N.; Pylypko, S.L.; Blower, R.J.; Ahmad, S.; Mansoor, M.; van Hoek, M.L. Francisella philomiragia Infection and Lethality
in Mammalian Tissue Culture Cell Models, Galleria mellonella, and BALB/c Mice. Front. Microbiol. 2016,7, 696. [CrossRef]
121.
Ozturk, S. Demonstration of the Efficacy of Curcumin on Carbapenem-Resistant Pseudomonas aeruginosa with Galleria mellonella
Larvae Model. Arch. Microbiol. 2022,204, 524. [CrossRef]
122.
Antoine, C.; Laforêt, F.; Blasdel, B.; Glonti, T.; Kutter, E.; Pirnay, J.P.; Mainil, J.; Delcenserie, V.; Thiry, D. Efficacy Assessment
of PEV2 Phage on Galleria mellonella Larvae Infected with a Pseudomonas aeruginosa Dog Otitis Isolate. Res. Vet. Sci.
2021
,136,
598–601. [CrossRef]
123.
Santos, T.A.; Scorzoni, L.; de Castro Santos, A.; Junqueira, J.C.; Anbinder, A.L. Galleria mellonella as an Experimental Model for
Studying Periodontopathogens. J. Indian Soc. Periodontol. 2020,24, 593–596. [CrossRef]
124.
Moraes, R.M.; Garcia, M.T.; Stossi, F.; de Barros, P.P.; Junqueira, J.C.; Anbinder, A.L. Effects of
α
and
β
-Adrenergic Signaling on
Innate Immunity and Porphyromonas Gingivalis Virulence in an Invertebrate Model. Virulence
2022
,13, 1614–1630. [CrossRef]
[PubMed]
125. Sowa-Jasiłek, A.; Zdybicka-Barabas, A.; St ˛aczek, S.; Wydrych, J.; Skrzypiec, K.; Mak, P.; Deryło, K.; Tchórzewski, M.; Cytry´nska,
M. Galleria mellonella Lysozyme Induces Apoptotic Changes in Candida albicans Cells. Microbiol. Res.
2016
,193, 121–131. [CrossRef]
126.
de Barros, P.P.; Rossoni, R.D.; de Camargo Ribeiro, F.; Silva, M.P.; de Souza, C.M.; Jorge, A.O.C.; Junqueira, J.C. Two Sporulated
Bacillus Enhance Immunity in Galleria mellonella Protecting against Candida albicans.Microb. Pathog.
2019
,132, 335–342. [CrossRef]
Microorganisms 2023,11, 1455 17 of 21
127.
Araújo, D.; Mil-Homens, D.; Henriques, M.; Silva, S. Anti-EFG1 2
0
-OMethylRNA Oligomer Inhibits Candida albicans Filamentation
and Attenuates the Candidiasis in Galleria mellonella.Mol. Ther. Nucleic Acids 2022,27, 517–523. [CrossRef]
128.
Kaskatepe, B.; Aslan Erdem, S.; Ozturk, S.; Safi Oz, Z.; Subasi, E.; Koyuncu, M.; Vlaini´c, J.; Kosalec, I. Antifungal and Anti-Virulent
Activity of Origanum majorana L. Essential Oil on Candida albicans and In Vivo Toxicity in the Galleria mellonella Larval Model.
Molecules 2022,27, 663. [CrossRef] [PubMed]
129.
Martins de Andrade, V.; Bardají, E.; Heras, M.; Ramu, V.G.; Junqueira, J.C.; Diane Dos Santos, J.; Castanho, M.A.R.B.; Conceição,
K. Antifungal and Anti-Biofilm Activity of Designed Derivatives from Kyotorphin. Fungal. Biol. 2020,124, 316–326. [CrossRef]
130.
Tagle-Olmedo, T.; Andrade-Pavón, D.; Martínez-Gamboa, A.; Gómez-García, O.; García-Sierra, F.; Hernández-Rodríguez, C.; Villa-
Tanaca, L. Inhibitors of DNA Topoisomerases I and II Applied to Candida dubliniensis Reduce Growth, Viability, the Generation of
Petite Mutants and Toxicity, While Acting Synergistically with Fluconazole. FEMS Yeast Res. 2021,21, foab023. [CrossRef]
131.
Özkan, S.; Coutts, R.H.A. Aspergillus fumigatus Mycovirus Causes Mild Hypervirulent Effect on Pathogenicity When Tested on
Galleria mellonella.Fungal. Genet. Biol. 2015,76, 20–26. [CrossRef]
132.
Curtis, A.; Walshe, K.; Kavanagh, K. Prolonged Subculturing of Aspergillus fumigatus on Galleria Extract Agar Results in Altered
Virulence and Sensitivity to Antifungal Agents. Cells 2023,12, 1065. [CrossRef] [PubMed]
133.
Eisenman, H.C.; Duong, R.; Chan, H.; Tsue, R.; McClelland, E.E. Reduced Virulence of Melanized Cryptococcus neoformans in
Galleria mellonella.Virulence 2014,5, 611–618. [CrossRef] [PubMed]
134.
Ali, M.F.; Tansie, S.M.; Shahan, J.R.; Seipelt-Thiemann, R.L.; McClelland, E.E. Serial Passage of Cryptococcus neoformans in Galleria
mellonella Results in Increased Capsule and Intracellular Replication in Hemocytes, but Not Increased Resistance to Hydrogen
Peroxide. Pathogens 2020,9, 732. [CrossRef] [PubMed]
135.
Trevijano-Contador, N.; Herrero-Fernández, I.; García-Barbazán, I.; Scorzoni, L.; Rueda, C.; Rossi, S.A.; García-Rodas, R.; Zaragoza,
O. Cryptococcus neoformans Induces Antimicrobial Responses and Behaves as a Facultative Intracellular Pathogen in the Non
Mammalian Model Galleria mellonella.Virulence 2015,6, 66–74. [CrossRef] [PubMed]
136.
Lim, W.; Konings, M.; Parel, F.; Eadie, K.; Strepis, N.; Fahal, A.; Verbon, A.; van de Sande, W.W.J. Inhibiting DHN- and DOPA-
Melanin Biosynthesis Pathway Increased the Therapeutic Value of Itraconazole in Madurella mycetomatis Infected Galleria mellonella.
Med. Mycol. 2022,60, myac003. [CrossRef]
137.
Sheehan, G.; Konings, M.; Lim, W.; Fahal, A.; Kavanagh, K.; van de Sande, W.W.J. Proteomic Analysis of the Processes Leading to
Madurella mycetomatis Grain Formation in Galleria mellonella Larvae. PLoS Negl. Trop. Dis. 2020,14, e0008190. [CrossRef]
138.
Eadie, K.; Parel, F.; Helvert-van Poppel, M.; Fahal, A.; van de Sande, W. Combining Two Antifungal Agents Does Not Enhance
Survival of Galleria mellonella Larvae Infected with Madurella mycetomatis.Trop. Med. Int. Health 2017,22, 696–702. [CrossRef]
139.
Champion, O.; Titball, R.; Bates, S. Standardization of G. mellonella Larvae to Provide Reliable and Reproducible Results in the
Study of Fungal Pathogens. JoF 2018,4, 108. [CrossRef]
140.
Constantino, M.; Christian, P.; Marina, C.F.; Williams, T. A Comparison of Techniques for Detecting Invertebrate iridescent Virus 6.
J. Virol. Methods 2001,98, 109–118. [CrossRef]
141.
Rossoni, R.D.; de Camargo Ribeiro, F.; Dos Santos, H.F.S.; Dos Santos, J.D.; de Sousa Oliveira, N.; Dutra, M.T.D.S.; de Lapena,
S.A.B.; Junqueira, J.C. Galleria mellonella as an Experimental Model to Study Human Oral Pathogens. Arch. Oral Biol.
2019
,101,
13–22. [CrossRef]
142.
Kaya, S.; Uçkan, F.; Er, A. Immunosuppressive Influence of Parasitoid Wasp Pimpla turionellae Calyx Fluid on Host Galleria
mellonella Cell-Mediated Immune Response and Hemocyte Viability. Bull. Entomol. Res.
2021
,112, 361–369. [CrossRef] [PubMed]
143.
El Haddad, L.; Angelidakis, G.; Clark, J.R.; Mendoza, J.F.; Terwilliger, A.L.; Chaftari, C.P.; Duna, M.; Yusuf, S.T.; Harb, C.P.; Stibich,
M.; et al. Genomic and Functional Characterization of Vancomycin-Resistant Enterococci-Specific Bacteriophages in the Galleria
mellonella Wax Moth Larvae Model. Pharmaceutics 2022,14, 1591. [CrossRef] [PubMed]
144.
Jeon, J.; Yong, D. Two Novel Bacteriophages Improve Survival in Galleria mellonella Infection and Mouse Acute Pneumonia
Models Infected with Extensively Drug-Resistant Pseudomonas aeruginosa.Appl. Environ. Microbiol.
2019
,85, e02900-18. [CrossRef]
[PubMed]
145.
Manohar, P.; Nachimuthu, R.; Lopes, B.S. The Therapeutic Potential of Bacteriophages Targeting Gram-Negative Bacteria Using
Galleria mellonella Infection Model. BMC Microbiol. 2018,18, 97. [CrossRef]
146.
Nale, J.Y.; Chutia, M.; Carr, P.; Hickenbotham, P.T.; Clokie, M.R.J. “Get in Early”; Biofilm and Wax Moth (Galleria mellonella)
Models Reveal New Insights into the Therapeutic Potential of Clostridium Difficile Bacteriophages. Front. Microbiol.
2016
,7, 1383.
[CrossRef]
147.
Sänger, P.-A.; Wagner, S.; Liebler-Tenorio, E.M.; Fuchs, T.M. Dissecting the Invasion of Galleria mellonella by Yersinia Enterocolitica
Reveals Metabolic Adaptations and a Role of a Phage Lysis Cassette in Insect Killing. PLoS Pathog.
2022
,18, e1010991. [CrossRef]
148.
Filippov, A.A.; Su, W.; Sergueev, K.V.; Kevorkian, R.T.; Snesrud, E.C.; Srijan, A.; He, Y.; Fouts, D.E.; Lurchachaiwong, W.; McGann,
P.T.; et al. Design of a Bacteriophage Cocktail Active against Shigella Species and Testing of Its Therapeutic Potential in Galleria
mellonella.Antibiotics 2022,11, 1659. [CrossRef]
149.
Li, Y.; Pu, M.; Han, P.; Li, M.; An, X.; Song, L.; Fan, H.; Chen, Z.; Tong, Y. Efficacy in Galleria mellonella Larvae and Application
Potential Assessment of a New Bacteriophage BUCT700 Extensively Lyse Stenotrophomonas maltophilia.Microbiol. Spectr.
2023
,11,
e04030-22. [CrossRef]
150.
Abbasifar, R.; Kropinski, A.M.; Sabour, P.M.; Chambers, J.R.; MacKinnon, J.; Malig, T.; Griffiths, M.W. Efficiency of Bacteriophage
Therapy against Cronobacter sakazakii in Galleria mellonella (Greater Wax Moth) Larvae. Arch. Virol.
2014
,159, 2253–2261. [CrossRef]
Microorganisms 2023,11, 1455 18 of 21
151.
Vergis, J.; Malik, S.; Pathak, R.; Kumar, M.; Ramanjaneya, S.; Kurkure, N.; Barbuddhe, S.B.; Rawool, D.B. Efficacy of Indolicidin,
CAMA, Lactoferricin (17–30) and Their Combination against Multi-Drug Resistant Enteroaggregative Escherichia coli.Int. J. Infect.
Dis. 2020,101, 11. [CrossRef]
152.
Kavanagh, K.; Sheehan, G. The Use of Galleria mellonella Larvae to Identify Novel Antimicrobial Agents against Fungal Species of
Medical Interest. JoF 2018,4, 113. [CrossRef] [PubMed]
153.
Trevijano-Contador, N.; Zaragoza, O. Immune Response of Galleria mellonella against Human Fungal Pathogens. JoF
2018
,5, 3.
[CrossRef] [PubMed]
154.
Ménard, G.; Rouillon, A.; Ghukasyan, G.; Emily, M.; Felden, B.; Donnio, P.-Y. Galleria mellonella Larvae as an Infection Model
to Investigate SRNA-Mediated Pathogenesis in Staphylococcus aureus.Front. Cell. Infect. Microbiol.
2021
,11, 631710. [CrossRef]
[PubMed]
155.
Junqueira, J.C. Galleria mellonella as a Model Host for Human Pathogens: Recent Studies and New Perspectives. Virulence
2012
,3,
474–476. [CrossRef] [PubMed]
156.
Chen, R.Y.; Keddie, B.A. Galleria mellonella (Lepidoptera: Pyralidae) Hemocytes Release Extracellular Traps That Confer Protection
Against Bacterial Infection in the Hemocoel. J. Insect Sci. 2021,21, 17. [CrossRef]
157.
Brinkmann, V.; Reichard, U.; Goosmann, C.; Fauler, B.; Uhlemann, Y.; Weiss, D.S.; Weinrauch, Y.; Zychlinsky, A. Neutrophil
Extracellular Traps Kill Bacteria. Science 2004,303, 1532–1535. [CrossRef]
158.
Browne, N.; Heelan, M.; Kavanagh, K. An Analysis of the Structural and Functional Similarities of Insect Hemocytes and
Mammalian Phagocytes. Virulence 2013,4, 597–603. [CrossRef]
159.
Altincicek, B.; Stötzel, S.; Wygrecka, M.; Preissner, K.T.; Vilcinskas, A. Host-Derived Extracellular Nucleic Acids Enhance Innate
Immune Responses, Induce Coagulation, and Prolong Survival upon Infection in Insects. J. Immunol.
2008
,181, 2705–2712.
[CrossRef]
160.
Wright, C.L.; Kavanagh, O. Galleria mellonella as a Novel In Vivo Model to Screen Natural Product-Derived Modulators of Innate
Immunity. Appl. Sci. 2022,12, 6587. [CrossRef]
161.
Bergin, D.; Reeves, E.P.; Renwick, J.; Wientjes, F.B.; Kavanagh, K. Superoxide Production in Galleria mellonella Hemocytes:
Identification of Proteins Homologous to the NADPH Oxidase Complex of Human Neutrophils. Infect. Immun.
2005
,73,
4161–4170. [CrossRef]
162.
Bismuth, H.D.; Brasseur, G.; Ezraty, B.; Aussel, L. Bacterial Genetic Approach to the Study of Reactive Oxygen Species Production
in Galleria mellonella During Salmonella Infection. Front. Cell. Infect. Microbiol. 2021,11, 640112. [CrossRef] [PubMed]
163.
Slepneva, I.A.; Glupov, V.V.; Sergeeva, S.V.; Khramtsov, V.V. EPR Detection of Reactive Oxygen Species in Hemolymph of Galleria
mellonella and Dendrolimus superans sibiricus (Lepidoptera) Larvae. Biochem. Biophys. Res. Commun.
1999
,264, 212–215. [CrossRef]
[PubMed]
164.
Wro´nska, A.K.; Kaczmarek, A.; Kazek, M.; Bogu´s, M.I. Infection of Galleria mellonella (Lepidoptera) Larvae with the Ento-
mopathogenic Fungus Conidiobolus coronatus (Entomophthorales) Induces Apoptosis of Hemocytes and Affects the Concentration
of Eicosanoids in the Hemolymph. Front. Physiol. 2022,12, 774086. [CrossRef] [PubMed]
165.
Pereira, T.; de Barros, P.; Fugisaki, L.; Rossoni, R.; Ribeiro, F.; de Menezes, R.; Junqueira, J.; Scorzoni, L. Recent Advances in the
Use of Galleria mellonella Model to Study Immune Responses against Human Pathogens. JoF 2018,4, 128. [CrossRef] [PubMed]
166.
Vilcinskas, A. ANTI-Infective Therapeutics from the Lepidopteran Model Host Galleria mellonella.CPD
2011
,17, 1240–1245.
[CrossRef] [PubMed]
167.
Kordaczuk, J.; Sułek, M.; Mak, P.; Zdybicka-Barabas, A.; ´
Smiałek, J.; Wojda, I. Cationic Protein 8 Plays Multiple Roles in Galleria
mellonella Immunity. Sci. Rep. 2022,12, 11737. [CrossRef]
168.
Gandra, R.M.; McCarron, P.; Viganor, L.; Fernandes, M.F.; Kavanagh, K.; McCann, M.; Branquinha, M.H.; Santos, A.L.S.; Howe,
O.; Devereux, M. In Vivo Activity of Copper(II), Manganese(II), and Silver(I) 1,10-Phenanthroline Chelates Against Candida
haemulonii Using the Galleria mellonella Model. Front. Microbiol. 2020,11, 470. [CrossRef]
169.
de Oliveira, F.E.; Rossoni, R.D.; de Barros, P.P.; Begnini, B.E.; Junqueira, J.C.; Jorge, A.O.C.; Leão, M.V.P.; de Oliveira, L.D.
Immunomodulatory Effects and Anti-Candida Activity of Lactobacilli in Macrophages and in Invertebrate Model of Galleria
mellonella.Microbial. Pathog. 2017,110, 603–611. [CrossRef]
170.
Rossoni, R.D.; Fuchs, B.B.; de Barros, P.P.; dos Santos Velloso, M.; Jorge, A.O.C.; Junqueira, J.C.; Mylonakis, E. Lactobacillus
paracasei Modulates the Immune System of Galleria mellonella and Protects against Candida albicans Infection. PLoS ONE
2017
,12,
e0173332. [CrossRef]
171.
de Melo, N.R.; Abdrahman, A.; Greig, C.; Mukherjee, K.; Thornton, C.; Ratcliffe, N.A.; Vilcinskas, A.; Butt, T.M. Myriocin
Significantly Increases the Mortality of a Non-Mammalian Model Host during Candida Pathogenesis. PLoS ONE
2013
,8, e78905.
[CrossRef]
172.
Yi, Y.; Wu, G.; Lv, J.; Li, M. Eicosanoids Mediate Galleria mellonella Immune Response to Hemocoel Injection of Entomopathogenic
Nematode Cuticles. Parasitol. Res. 2016,115, 597–608. [CrossRef] [PubMed]
173.
Mc Namara, L.; Dolan, S.K.; Walsh, J.M.D.; Stephens, J.C.; Glare, T.R.; Kavanagh, K.; Griffin, C.T. Oosporein, an Abundant
Metabolite in Beauveria caledonica, with a Feedback Induction Mechanism and a Role in Insect Virulence. Fungal. Biol.
2019
,123,
601–610. [CrossRef] [PubMed]
Microorganisms 2023,11, 1455 19 of 21
174.
Brivio, M.F.; Toscano, A.; De Pasquale, S.M.; De Lerma Barbaro, A.; Giovannardi, S.; Finzi, G.; Mastore, M. Surface Protein
Components from Entomopathogenic Nematodes and Their Symbiotic Bacteria: Effects on Immune Responses of the Greater
Wax Moth, Galleria mellonella (Lepidoptera: Pyralidae): Immune-Depressive Role of Surface Components of Nematocomplexes.
Pest. Manag. Sci. 2018,74, 2089–2099. [CrossRef] [PubMed]
175.
Fiolka, M.J. Immunosuppressive Effect of Cyclosporin A on Insect Humoral Immune Response. J. Invertebr. Pathol.
2008
,98,
287–292. [CrossRef] [PubMed]
176.
Vilcinskas, A.; Jegorov, A.; Landa, Z.; Götz, P.; Matha, V. Effects of Beauverolide L and Cyclosporin A on Humoral and Cellular
Immune Response of the Greater Wax Moth, Galleria mellonella.Comp. Biochem. Physiol. Part Pharmacol. Toxicol. Endocrinol.
1999
,
122, 83–92. [CrossRef]
177.
Shi, H.; Zeng, H.; Yang, X.; Liu, Z.; Qiu, D. An Insecticidal Protein from Xenorhabdus ehlersii Stimulates the Innate Immune
Response in Galleria mellonella.World J. Microbiol. Biotechnol. 2013,29, 1705–1711. [CrossRef]
178.
Sheehan, G.; Margalit, A.; Sheehan, D.; Kavanagh, K. Proteomic Profiling of Bacterial and Fungal Induced Immune Priming in
Galleria mellonella Larvae. J. Insect Physiol. 2021,131, 104213. [CrossRef]
179.
dos Santos, A.F.; de Almeida, D.R.Q.; Terra, L.F.; Baptista, M.S.; Labriola, L. Photodynamic Therapy in Cancer Treatment—An
Update Review. J. Cancer Metastasis Treat. 2019,5, 25. [CrossRef]
180.
Patiño-Márquez, I.A.; Patiño-González, E.; Hernández-Villa, L.; Ortíz-Reyes, B.; Manrique-Moreno, M. Identification and
Evaluation of Galleria mellonella Peptides with Antileishmanial Activity. Anal. Biochem. 2018,546, 35–42. [CrossRef]
181.
Mowlds, P.; Barron, A.; Kavanagh, K. Physical Stress Primes the Immune Response of Galleria mellonella Larvae to Infection by
Candida albicans.Microbes Infect. 2008,10, 628–634. [CrossRef]
182.
Fallon, J.P.; Troy, N.; Kavanagh, K. Pre-Exposure of Galleria mellonella Larvae to Different Doses of Aspergillus Fumigatus Conidia
Causes Differential Activation of Cellular and Humoral Immune Responses. Virulence 2011,2, 413–421. [CrossRef] [PubMed]
183.
Brivio, M.F.; Mastore, M.; Nappi, A.J. A Pathogenic Parasite Interferes with Phagocytosis of Insect Immunocompetent Cells. Dev.
Comp. Immunol. 2010,34, 991–998. [CrossRef] [PubMed]
184.
Shaik, H.A.; Sehnal, F. Hemolin Expression in the Silk Glands of Galleria mellonella in Response to Bacterial Challenge and Prior to
Cell Disintegration. J. Insect Physiol. 2009,55, 781–787. [CrossRef]
185.
Feng, P.; Shang, Y.; Cen, K.; Wang, C. Fungal Biosynthesis of the Bibenzoquinone Oosporein to Evade Insect Immunity. Proc. Natl.
Acad. Sci. USA 2015,112, 11365–11370. [CrossRef] [PubMed]
186.
Kelly, J.; Kavanagh, K. Caspofungin Primes the Immune Response of the Larvae of Galleria mellonella and Induces a Non-Specific
Antimicrobial Response. J. Med. Microbiol. 2011,60, 189–196. [CrossRef] [PubMed]
187.
Torres, M.P.; Entwistle, F.; Coote, P.J. Effective Immunosuppression with Dexamethasone Phosphate in the Galleria mellonella
Larva Infection Model Resulting in Enhanced Virulence of Escherichia coli and Klebsiella kneumoniae.Med. Microbiol. Immunol.
2016,205, 333–343. [CrossRef]
188.
Liu, H.; Zeng, H.; Yao, Q.; Yuan, J.; Zhang, Y.; Qiu, D.; Yang, X.; Yang, H.; Liu, Z. Steinernema Glaseri Surface Enolase: Molecular
Cloning, Biological Characterization, and Role in Host Immune Suppression. Mol. Biochem. Parasitol.
2012
,185, 89–98. [CrossRef]
189.
Wu, G.; Liu, Y.; Ding, Y.; Yi, Y. Ultrastructural and Functional Characterization of Circulating Hemocytes from Galleria mellonella
Larva: Cell Types and Their Role in the Innate Immunity. Tissue Cell 2016,48, 297–304. [CrossRef]
190.
Kazek, M.; Kaczmarek, A.; Wro´nska, A.K.; Bogu´s, M.I. Conidiobolus coronatus Induces Oxidative Stress and Autophagy Response
in Galleria mellonella Larvae. PLoS ONE 2020,15, e0228407. [CrossRef]
191.
Szurpnicka, A.; Wro´nska, A.K.; Bus, K.; Kozi ´nska, A.; Jabłczy´nska, R.; Szterk, A.; Lubelska, K. Phytochemical Screening and
Effect of Viscum album L. on Monoamine Oxidase A and B Activity and Serotonin, Dopamine and Serotonin Receptor 5-HTR1A
Levels in Galleria mellonealla (Lepidoptera). J. Ethnopharmacol. 2022,298, 115604. [CrossRef]
192.
Spagnul, C.; Turner, L.C.; Boyle, R.W. Immobilized Photosensitizers for Antimicrobial Applications. J. Photochem. Photobiol. Biol.
2015,150, 11–30. [CrossRef] [PubMed]
193.
Dolmans, D.E.J.G.J.; Fukumura, D.; Jain, R.K. Photodynamic Therapy for Cancer. Nat. Rev. Cancer
2003
,3, 380–387. [CrossRef]
[PubMed]
194. Konopka, K.; Goslinski, T. Photodynamic Therapy in Dentistry. J. Dent. Res. 2007,86, 694–707. [CrossRef]
195.
Dougherty, T.J.; Gomer, C.J.; Henderson, B.W.; Jori, G.; Kessel, D.; Korbelik, M.; Moan, J.; Peng, Q. Photodynamic Therapy. J. Natl.
Cancer Inst. 1998,90, 889–905. [CrossRef]
196.
Fonseca, L.L.; Durães, C.P.; da Silva Menezes, A.S.; Tabosa, A.T.L.; Barbosa, C.U.; de Paulo Santiago Filho, A.; de Paula Souza,
D.P.S.; Guimarães, V.H.D.; Santos, S.H.S.; de Paula, A.M.B.; et al. Comparison between Two Antimicrobial Photodynamic Therapy
Protocols for Oral Candidiasis in Patients Undergoing Treatment for Head and Neck Cancer: A Two-Arm, Single-Blind Clinical
Trial. Photodiagnosis Photodyn. Ther. 2022,39, 102983. [CrossRef] [PubMed]
197.
Shetty, B.; Ali, D.; Ahmed, S.; Ibraheem, W.I.; Preethanath, R.S.; Vellappally, S.; Divakar, D.D. Role of Antimicrobial Photodynamic
Therapy in Reducing Subgingival Oral Yeasts Colonization in Patients with Peri-Implant Mucositis. Photodiagnosis. Photodyn.
Ther. 2022,38, 102803. [CrossRef] [PubMed]
198.
Alves-Silva, E.G.; Arruda-Vasconcelos, R.; Louzada, L.M.; de-Jesus-Soares, A.; Ferraz, C.C.R.; Almeida, J.F.A.; Marciano, M.A.;
Steiner-Oliveira, C.; Santos, J.M.M.; Gomes, B.P. Effect of Antimicrobial Photodynamic Therapy on the Reduction of Bacteria and
Virulence Factors in Teeth with Primary Endodontic Infection. Photodiagnosis. Photodyn. Ther. 2023,41, 103292. [CrossRef]
Microorganisms 2023,11, 1455 20 of 21
199.
Yilmaz, A.; Ozkiraz, S.; Akcan, A.B.; Canpolat, M. Low-Cost Home-Use Light-Emitting-Diode Phototherapy as an Alternative to
Conventional Methods. J. Trop. Pediatr. 2015,61, 113–118. [CrossRef]
200.
Stockett, M.H.; Musbat, L.; Kjær, C.; Houmøller, J.; Toker, Y.; Rubio, A.; Milne, B.F.; Brøndsted Nielsen, S. The Soret Absorption
Band of Isolated Chlorophyll a and b Tagged with Quaternary Ammonium Ions. Phys. Chem. Chem. Phys.
2015
,17, 25793–25798.
[CrossRef]
201.
Tortik, N.; Steinbacher, P.; Maisch, T.; Spaeth, A.; Plaetzer, K. A Comparative Study on the Antibacterial Photodynamic Efficiency
of a Curcumin Derivative and a Formulation on a Porcine Skin Model. Photochem. Photobiol. Sci. 2016,15, 187–195. [CrossRef]
202.
ˇ
Cernáková, L.; Light, C.; Salehi, B.; Rogel-Castillo, C.; Victoriano, M.; Martorell, M.; Sharifi-Rad, J.; Martins, N.; Rodrigues, C.F.
Novel Therapies for Biofilm-Based Candida Spp. Infections. Adv. Exp. Med. Biol. 2019,1214, 93–123. [CrossRef]
203.
Ribeiro, M.; Gomes, I.B.; Saavedra, M.J.; Simões, M. Photodynamic Therapy and Combinatory Treatments for the Control of
Biofilm-Associated Infections. Lett. Appl. Microbiol. 2022,75, 548–564. [CrossRef] [PubMed]
204.
Štefánek, M.; ˇ
Cernáková, L.; Dekkerová, J.; Bujdáková, H. Photodynamic Inactivation Effectively Eradicates Candida auris Biofilm
despite Its Interference with the Upregulation of CDR1 and MDR1 Efflux Genes. J. Fungi. 2022,8, 1137. [CrossRef]
205.
Wimmer, A.; Glueck, M.; Ckurshumova, W.; Liu, J.; Fefer, M.; Plaetzer, K. Breaking the Rebellion: Photodynamic Inactivation
against Erwinia Amylovora Resistant to Streptomycin. Antibiotics 2022,11, 544. [CrossRef] [PubMed]
206.
Fekrirad, Z.; Kashef, N.; Arefian, E. Photodynamic Inactivation Diminishes Quorum Sensing-Mediated Virulence Factor Produc-
tion and Biofilm Formation of Serratia marcescens.World J. Microbiol. Biotechnol. 2019,35, 191. [CrossRef]
207.
Zangirolami, A.C.; Inada, N.M.; Bagnato, V.S.; Blanco, K.C. Biofilm Destruction on Endotracheal Tubes by Photodynamic
Inactivation. Infect. Disord Drug Targets 2018,18, 218–223. [CrossRef]
208.
Amorim, C.F.; Iglesias, B.A.; Pinheiro, T.R.; Lacerda, L.E.; Sokolonski, A.R.; Pedreira, B.O.; Moreira, K.S.; Burgo, T.A.L.; Meyer,
R.; Azevedo, V.; et al. Photodynamic Inactivation of Different Candida Species and Inhibition of Biofilm Formation Induced by
Water-Soluble Porphyrins. Photodiagnosis. Photodyn. Ther. 2023,42, 103343. [CrossRef]
209.
Merigo, E.; Conti, S.; Ciociola, T.; Fornaini, C.; Polonelli, L.; Lagori, G.; Manfredi, M.; Vescovi, P. Effect of Different Wavelengths
and Dyes on Candida albicans: In Vivo Study Using Galleria mellonella as an Experimental Model. Photodiagnosis Photodyn. Ther.
2017,18, 34–38. [CrossRef]
210.
Paziani, M.H.; Tonani, L.; de Menezes, H.D.; Bachmann, L.; Wainwright, M.; Braga, G.Ú.L.; von Zeska Kress, M.R. Antimicrobial
Photodynamic Therapy with Phenothiazinium Photosensitizers in Non-Vertebrate Model Galleria mellonella Infected with
Fusarium Keratoplasticum and Fusarium Moniliforme. Photodiagnosis Photodyn. Ther. 2019,25, 197–203. [CrossRef]
211.
Sanches, C.V.G.; de Cássia Orlandi Sardi, J.; Terada, R.S.S.; Lazarini, J.G.; Freires, I.A.; Polaquini, C.R.; Torrezan, G.S.; Regasini, L.O.;
Fujimaki, M.; Rosalen, P.L. Diacetylcurcumin: A New Photosensitizer for Antimicrobial Photodynamic Therapy in Streptococcus
mutans Biofilms. Biofouling 2019,35, 340–349. [CrossRef]
212.
Garcez, A.S.; Kaplan, M.; Jensen, G.J.; Scheidt, F.R.; Oliveira, E.M.; Suzuki, S.S. Effects of Antimicrobial Photodynamic Therapy
on Antibiotic-Resistant Escherichia coli.Photodiagnosis Photodyn. Ther. 2020,32, 102029. [CrossRef]
213.
Chibebe Junior, J.; Sabino, C.P.; Tan, X.; Junqueira, J.C.; Wang, Y.; Fuchs, B.B.; Jorge, A.O.; Tegos, G.P.; Hamblin, M.R.; Mylonakis,
E. Selective Photoinactivation of Candida albicans in the Non-Vertebrate Host Infection Model Galleria mellonella.BMC Microbiol.
2013,13, 217. [CrossRef]
214.
ˇ
Cernáková, L.; Chupáˇcová, J.; Židlíková, K.; Bujdáková, H. Effectiveness of the Photoactive Dye Methylene Blue versus
Caspofungin on the Candida Parapsilosis Biofilm in Vitro and Ex Vivo. Photochem. Photobiol. 2015,91, 1181–1190. [CrossRef]
215.
CRZP—Detail Kniha. Available online: https://opac.crzp.sk/?fn=detailBiblioFormChildA2KPL&sid=26764B43C57DF4A5B2
DA947C556C&seo=CRZP-detail-kniha (accessed on 27 April 2023).
216.
dos Santos, J.D.; de Alvarenga, J.A.; Rossoni, R.D.; García, M.T.; Moraes, R.M.; Anbinder, A.L.; Cardoso Jorge, A.O.; Junqueira,
J.C. Immunomodulatory Effect of Photodynamic Therapy in Galleria mellonella Infected with Porphyromonas Gingivalis. Microb.
Pathog. 2017,110, 507–511. [CrossRef] [PubMed]
217.
Marques Meccatti, V.; de Souza Moura, L.; Guerra Pinto, J.; Ferreira-Strixino, J.; Abu Hasna, A.; Alves Figueiredo-Godoi, L.M.;
Campos Junqueira, J.; Marcucci, M.C.; de Paula Ramos, L.; Carvalho, C.A.T.; et al. Curcuma Longa L. Extract and Photodynamic
Therapy Are Effective against Candida Spp. and Do Not Show Toxicity In Vivo. Int. J. Dent.
2022
,2022, 5837864. [CrossRef]
[PubMed]
218.
Plaetzer, K.; Krammer, B.; Berlanda, J.; Berr, F.; Kiesslich, T. Photophysics and Photochemistry of Photodynamic Therapy:
Fundamental Aspects. Lasers Med. Sci. 2009,24, 259–268. [CrossRef]
219.
Issa, M.C.A.; Manela-Azulay, M. Terapia Fotodinâmica: Revisão Da Literatura e Documentação Iconográfica. An. Bras. Dermatol.
2010,85, 501–511. [CrossRef]
220.
Allison, R.R.; Downie, G.H.; Cuenca, R.; Hu, X.-H.; Childs, C.J.; Sibata, C.H. Photosensitizers in Clinical PDT. Photodiagnosis
Photodyn. Ther. 2004,1, 27–42. [CrossRef] [PubMed]
221.
Meisel, P.; Kocher, T. Photodynamic Therapy for Periodontal Diseases: State of the Art. J. Photochem. Photobiol. Biol.
2005
,79,
159–170. [CrossRef] [PubMed]
222.
Grizante Barião, P.H.; Tonani, L.; Brancini, G.T.P.; Nascimento, E.; Braga, G.Ú.L.; Wainwright, M.; von Zeska Kress, M.R. In Vitro
and in Vivo Photodynamic Efficacies of Novel and Conventional Phenothiazinium Photosensitizers against Multidrug-Resistant
Candida auris.Photochem. Photobiol. Sci. 2022,21, 1807–1818. [CrossRef]
Microorganisms 2023,11, 1455 21 of 21
223.
Huang, X.; Xu, M.; Pan, W.; Wang, M.; Wu, X.; Dai, S.; Li, L.; Zeng, K. Antimicrobial and Immunomodulatory Responses of
Photodynamic Therapy in Galleria mellonella Model. BMC Microbiol. 2020,20, 196. [CrossRef]
224.
de França, B.M.; Ghasemishahrestani, Z.; de Souza, G.F.M.; da Silva, R.N.; Queiroz, D.D.; Pierre, M.B.R.; Pereira, M.D.; Forero,
J.S.B.; Corrêa, R.J. In Vitro Studies of Antitumor Effect, Toxicity/Cytotoxicity and Skin Permeation/Retention of a Green
Fluorescence Pyrene-Based Dye for PDT Application. Photochem. Photobiol. 2021,97, 408–415. [CrossRef] [PubMed]
225.
Rigotto Caruso, G.; Tonani, L.; Marcato, P.D.; von Zeska Kress, M.R. Phenothiazinium Photosensitizers Associated with Silver
Nanoparticles in Enhancement of Antimicrobial Photodynamic Therapy. Antibiotics 2021,10, 569. [CrossRef] [PubMed]
226.
Malacarne, M.C.; Mastore, M.; Gariboldi, M.B.; Brivio, M.F.; Caruso, E. Preliminary Toxicity Evaluation of a Porphyrin Photosen-
sitizer in an Alternative Preclinical Model. Int. J. Mol. Sci. 2023,24, 3131. [CrossRef] [PubMed]
227. Ghorbani, J.; Rahban, D.; Aghamiri, S.; Teymouri, A.; Bahador, A. Photosensitizers in Antibacterial Photodynamic Therapy: An
Overview. Laser Ther. 2018,27, 293–302. [CrossRef]
228.
Glueck, M.; Hamminger, C.; Fefer, M.; Liu, J.; Plaetzer, K. Save the Crop: Photodynamic Inactivation of Plant Pathogens I: Bacteria.
Photochem. Photobiol. Sci. 2019,18, 1700–1708. [CrossRef]
229.
Krüger, M.; Richter, P.; Strauch, S.; Nasir, A.; Burkovski, A.; Antunes, C.; Meißgeier, T.; Schlücker, E.; Schwab, S.; Lebert, M. What
an Escherichia coli Mutant Can Teach Us About the Antibacterial Effect of Chlorophyllin. Microorganisms
2019
,7, 59. [CrossRef]
[PubMed]
230.
Juzeniene, A.; Juzenas, P.; Ma, L.-W.; Iani, V.; Moan, J. Effectiveness of Different Light Sources for 5-Aminolevulinic Acid
Photodynamic Therapy. Lasers Med. Sci. 2004,19, 139–149. [CrossRef]
231.
Hasenleitner, M.; Plaetzer, K. In the Right Light: Photodynamic Inactivation of Microorganisms Using a LED-Based Illumination
Device Tailored for the Antimicrobial Application. Antibiotics 2019,9, 13. [CrossRef]
232.
Pereira Gonzales, F.; Maisch, T. Photodynamic Inactivation for Controlling Candida albicans Infections. Fungal. Biol.
2012
,116, 1–10.
[CrossRef]
233.
Garcez, A.S.; Hamblin, M.R. Methylene Blue and Hydrogen Peroxide for Photodynamic Inactivation in Root Canal—A New
Protocol for Use in Endodontics. Eur. Endod. J. 2017,2, 29. [CrossRef] [PubMed]
234.
Bispo, M.; Anaya-Sanchez, A.; Suhani, S.; Raineri, E.J.M.; López-Álvarez, M.; Heuker, M.; Szyma´nski, W.; Romero Pastrana, F.;
Buist, G.; Horswill, A.R.; et al. Fighting Staphylococcus aureus Infections with Light and Photoimmunoconjugates. JCI Insight
2020
,
5, e139512. [CrossRef] [PubMed]
Disclaimer/Publisher’s Note:
The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.
Article
Full-text available
In recent years, there has been a considerable increasing interest in the use of the greater wax moth Galleria mellonella as an animal model. In vivo pharmacological tests, concerning the efficacy and the toxicity of novel compounds are typically performed in mammalian models. However, the use of the latter is costly, laborious and requires ethical approval. In this context, G. mellonella larvae can be considered a valid option due to their greater ease of use and the absence of ethical rules. Furthermore, it has been demonstrated that the immune system of these invertebrates has similarity with the one of mammals, thus guaranteeing the reliability of this in vivo model, mainly in the microbiological field. To better develop the full potential of this model, we present a novel approach to characterize the hemocyte population from G. mellonella larvae and to highlight the immuno modulation upon infection and treatments. Our approach is based on the detection in isolated hemocytes from G. mellonella hemolymph of cell membrane markers typically expressed by human immune cells upon inflammation and infection, for instance CD14, CD44, CD80, CD163 and CD200. This method highlights the analogies between G. mellonella larvae and humans. Furthermore, we provide an innovative tool to perform pre-clinical evaluations of the efficacy of antimicrobial compounds in vivo to further proceed with clinical trials and support drug discovery campaigns.
Article
Full-text available
Aspergillus fumigatus is an environmental saprophyte and opportunistic fungal pathogen of humans. The aim of the work presented here was to examine the effect of serially subculturing A. fumigatus on agar generated from Galleria mellonella larvae in order to characterize the alterations in the phenotypes that might occur. The passaged strains showed alterations in virulence, antifungal susceptibility, and in protein abundances that may indicate adaptation after 25 passages over 231 days on Galleria extract agar. Passaged strains demonstrated reduced virulence in G. mellonella larvae and increased tolerance to hemocyte-mediated killing, hydrogen peroxide, itraconazole, and amphotericin B. A label-free proteomic analysis of control and passaged A. fumigatus strains revealed a total of 3329 proteins, of which 1902 remained following filtration, and 32 proteins were statistically significant as well as differentially abundant. Proteins involved in the response to oxidative stress were altered in abundance in the passaged strain and included (S)-S-oxide reductase (+2.63-fold), developmental regulator FlbA (+2.27-fold), and histone H2A.Z (−1.82-fold). These results indicate that the prolonged subculturing of A. fumigatus on Galleria extract agar results in alterations in the susceptibility to antifungal agents and in the abundance of proteins associated with the oxidative stress response. The phenomenon may be a result of selection for survival in adverse conditions and highlight how A. fumigatus may adapt to tolerate the pulmonary immune response in cases of human infection.
Article
Full-text available
Galleria mellonella (greater wax moth) larvae are used widely as surrogate infectious disease models, due to ease of use and the presence of an innate immune system functionally similar to that of vertebrates. Here we review G. mellonella-human intracellular bacteria pathogen infection models from the genera Burkholderia, Coxiella, Francisella, Listeria, and Mycobacterium. For all genera, G. mellonella use has increased understanding of host-bacterial interactive biology, particularly through studies comparing the virulence of closely related species and/or wild-type versus mutant pairs. In many cases, virulence in G. mellonella mirrors that found in mammalian infection models, although it is unclear whether the pathogenic mechanisms are the same. The use of G. mellonella larvae has speeded up in vivo efficacy and toxicity testing of novel antimicrobials to treat infections caused by intracellular bacteria: an area that will expand since the FDA no longer requires animal testing for licensure. Further use of G. mellonella-intracellular bacteria infection models will be driven by advances in G. mellonella genetics, imaging, metabolomics, proteomics, and transcriptomic methodologies, alongside the development and accessibility of reagents to quantify immune markers, all of which will be underpinned by a fully annotated genome.
Article
Full-text available
The first research on the insect Galleria mellonella was published 85 years ago, and the larva is now widely used as a model to study infections caused by bacterial and fungal pathogens, for screening new antimicrobials, to study the adjacent immune response in co-infections or in host-pathogen interaction, as well as in a toxicity model. The immune system of the G. mellonella model shows remarkable similarities with mammals. Furthermore, results from G. mellonella correlate positively with mammalian models and with other invertebrate models. Unlike other invertebrate models, G. mellonella can withstand temperatures of 37 °C, and its handling and experimental procedures are simpler. Despite having some disadvantages, G. mellonella is a virtuous in vivo model to be used in preclinical studies, as an intermediate model between in vitro and mammalian in vivo studies, and is a great example on how to apply the bioethics principle of the 3Rs (Replacement, Reduction, and Refinement) in animal experimentation. This review aims to discuss the progress of the G. mellonella model, highlighting the key aspects of its use, including experimental design considerations and the necessity to standardize them. A different score in the “cocoon” category included in the G. mellonella Health Index Scoring System is also proposed.
Article
Full-text available
Helcococcus ovis (H. ovis) can cause disease in a broad range of animal hosts, including humans, and has been described as an emerging bacterial pathogen in bovine metritis, mastitis, and endocarditis. In this study, we developed an infection model that showed H. ovis can proliferate in the hemolymph and induce dose-dependent mortality in the invertebrate model organism Galleria mellonella (G. mellonella). We applied the model and identified H. ovis isolates with attenuated virulence originating from the uterus of a healthy post-partum dairy cow (KG38) and hypervirulent isolates (KG37, KG106) originating from the uterus of cows with metritis. Medium virulence isolates were also isolated (KG36, KG104) from the uterus of cows with metritis. A major advantage of this model is that a clear differentiation in induced mortality between H. ovis isolates was detected in just 48 h, resulting in an effective infection model able to identify virulence differences between H. ovis isolates with a short turnaround time. Histopathology showed G. mellonella employs hemocyte-mediated immune responses to H. ovis infection, which are analogous to the innate immune response in cows. In summary, G. mellonella can be used as an invertebrate infection model for the emerging multi-host pathogen Helcococcus ovis.
Article
Full-text available
Purpose: This study aimed to evaluate and compare the presence of genes related to surface proteins between isolates of Streptococcus pneumoniae from healthy carriers (HC) and invasive pneumococcal disease (IPD) with a particular focus on serotype 19A. Methods: The presence of these genes was identified by real-time PCR. Subsequently, we employed the Galleria mellonella larval infection model to study their effect on pathogenicity in vivo. Results: The percentage of selected virulence genes was similar between the HC and IPD groups (p > 0.05), and the genes lytA, nanB, pavA, pcpA, phtA, phtB, phtE, rrgA, and sipA were all present in both groups. However, the virulence profile of the isolates differed individually between HC and IPD groups. The highest lethality in G. mellonella was for IPD isolates (p < 0.01), even when the virulence profile was the same as compared to the HC isolates or when the nanA, pspA, pspA-fam1, and pspC genes were not present. Conclusions: The occurrence of the investigated virulence genes was similar between HC and IPD S. pneumoniae serotype 19A groups. However, the IPD isolates showed a higher lethality in the alternative G. mellonella model than the HC isolates, regardless of the virulence gene composition, indicating that other virulence factors may play a decisive role in virulence. Currently, this is the first report using the in vivo G. mellonella model to study the virulence of clinical isolates of S. pneumoniae.
Article
Full-text available
Abstract: In photodynamic therapy (PDT), a photosensitizer (PS) excited with a specific wavelength, and in the presence of oxygen, gives rise to photochemical reactions that lead to cell damage. Over the past few years, larval stages of the G. mellonella moth have proven to be an excellent alternative animal model for in vivo toxicity testing of novel compounds and virulence testing. In this article, we report a series of preliminary studies on G. mellonella larvae to evaluate the photoinduced stress response by a porphyrin (PS) (TPPOH). The tests performed evaluated PS toxicity on larvae and cytotoxicity on hemocytes, both in dark conditions and following PDT. Cellular uptake was also evaluated by fluorescence and flow cytometry. The results obtained demonstrate how the administration of PS and subsequent irradiation of larvae affects not only larvae survival rate, but also immune system cells. It was also possible to verify PS's uptake and uptake kinetics in hemocytes, observing a maximum peak at 8 h. Given the results obtained in these preliminary tests, G. mellonella appears to be a promising model for preclinical PS tests.
Article
Background: Candida spp. is the main fungal genus related to infections in humans, and its treatment has become a challenge due to the production of biofilm and its resistance/multi-resistance profile to conventional antifungals. Antimicrobial photodynamic therapy stands out as a treatment characterized by a broad spectrum of antimicrobial action, being able to induce oxidative stress in pathogens, and porphyrins are photosensitizers with high selectivity to pathogens. Thus, this work aimed to analyze the photoinactivation of different species of Candida by two cationic (4-H2TMeP+ and 3-H2TMeP+) and one anionic (4-H2TPSP‒) porphyrins. Materials and methods: Microdilution assays were performed to determine the MIC100, with subsequent determination of MFC100. Determination of oxidative species was done through the use of scavengers, while biofilm morphological features were investigated using the atomic force microscopy. Results: Cationic porphyrins were significantly efficient in inactivating Candida albicans and non-albicans species with 100% growth inhibition and fungicidal activity (MFC100/MIC100 ≤ 4.0). The cationic porphyrins were also able to interfere in Candida spp biofilm formation. The photo-oxidative mechanism activated by 3-H2TMeP+ in Candida spp. is concurrent with the production of singlet oxygen and oxygen radical species. In the AFM analysis, 3-H2TMeP+ was able to reduce yeast adhesion to the surface. Conclusions: Cationic porphyrins can photo-inactivate different species of Candida in both planktonic and biofilm-associated forms, and reduce the adhesion of these fungi to the surface.
Article
Objective: To evaluate the effectiveness of supplemental photodynamic therapy for improving the bacterial removal and the levels of lipopolysaccharide (LPS) and lipoteichoic acid (LTA) by conducting a clinical trial. Methodology: Twenty-four root canals with pulp necrosis and periapical lesion were selected and randomly divided into conventional group using endodontic treatment with chemo-mechanical preparation (CMP) alone (n=12) and a group using antimicrobial photodynamic therapy (aPDT) after CMP (n=12). The samples were collected before and after CMP (conventional group) and after photodynamic therapy (aPDT group). A photosensitizer (0.005% methylene blue) was applied to the root canal for 3 minutes after CMP, whereas aPDT was performed by using a red laser with a power of 30Mw and energy density of 9J/cm2 for 90 s per root canal. Culture technique was performed to determine the bacterial colony forming units. LPS and LTA levels were quantified by using limulus amoebocyte lysate (LAL) assay and enzyme-linked immunosorbent assay (ELISA), respectively. Results: All samples showed growth of viable bacteria on Fastidious Anaerobe agar (FAA), with an average of 5.19 × 105 CFU/ mL. CMP was effective in decreasing viable bacteria (P<0.05), whereas there was a significant decrease (P<0.05) in the samples treated with aPDT compared to those submitted to CMP. LPS and LTA were detected in all initial samples, with mean values of 20.561 EU/mL and 430.91 pg/mL, respectively. Both CMP and aPDT groups significantly decreased the levels of LPS and LTA (P<0.05), with a statistical difference between the groups regarding aPDT (P<0.05). Conclusion: Photodynamic therapy as an adjunct to CMP proved to be effective in improving the bacterial disinfection and reducing the LPS and LTA levels in teeth with primary endodontic infection.
Article
Streptococcus mutans is a key pathogen associated with dental caries and is often implicated in infective endocarditis. This organism forms robust biofilms on tooth surfaces and can use collagen binding proteins (CBPs) to efficiently colonize collagenous substrates including dentin and heart valves. One of the best characterized CBPs of S. mutans is Cnm which contributes to adhesion and invasion of oral epithelial and heart endothelial cells. These virulence properties were subsequently linked to post-translational modification (PTM) of the Cnm threonine-rich repeat region by the Pgf glycosylation machinery, which consists of four enzymes: PgfS, PgfM1, PgfE and PgfM2. Inactivation of the S. mutans pgf genes leads to decreased collagen binding, reduced invasion of human coronary artery endothelial cells and attenuated virulence in the Galleria mellonella invertebrate model. The present study aimed to better understand Cnm glycosylation and to characterize the predicted 4-epimerase, PgfE. Using a truncated Cnm variant containing only two threonine-rich repeats, mass spectrometric analysis revealed extensive glycosylation with HexNAc2. Compositional analysis, complemented with lectin blotting, identified the HexNAc2 moieties as GlcNAc and GalNAc. Comparison of PgfE with the other S. mutans 4-epimerase GalE through structural modeling, NMR, and capillary electrophoresis, demonstrated that GalE is a UDP-Glc-4-epimerase while PgfE is a GlcNAc-4-epimerase. While PgfE exclusively participates in protein O-glycosylation, we found that GalE affects galactose metabolism and cell division. This study further emphasizes the importance of O-linked protein glycosylation and carbohydrate metabolism in S. mutans and identifies the PTM modifications of the key CBP, Cnm.