You are on page 1of 13

Journal of Applied Microbiology ISSN 1364-5072

REVIEW ARTICLE

Diagnosis of biofilm infections: current methods used,


challenges and perspectives for the future
N.B.S. Silva1 € der3
, L.A. Marques2 and D.D.B. Ro
1 Applied Immunology and Parasitology, Institute of Biomedical Sciences, Federal University of Uberl^ andia, Uberl^
andia, Minas Gerais, Brazil
2 Health Sciences, Medical School, Federal University of Uberl^ andia, Uberl^
andia, Minas Gerais, Brazil
3 Institute of Biomedical Sciences, Federal University of Uberl^
andia, Uberl^
andia, Minas Gerais, Brazil

Keywords Summary
clinical practice, diagnosis of biofilm, in vitro The diagnosis of biofilms continues to be a challenge, and there is no
methods, in vivo methods, MALDI-TOF MS,
standardized protocol for such a diagnosis in clinical practice. In addition,
omics analysis.
some proposed methodologies are expensive to require significant amounts of
Correspondence time and a high number of trained staff, making them impracticable for
Nagela B.S. Silva, Applied Immunology and clinical practice. In recent years, mass spectrophotometry/matrix-assisted laser
Parasitology, Institute of Biomedical Sciences, desorption ionization time of flight (MALDI-TOF) has been applied it in
Federal University of Uberl^andia, Av. Ama- biofilm studies. However, due to several problems and limitations of the
zonas, S/N - Umuarama, Uberl^andia, Minas technique, MALDI-TOF is far from being the gold standard for identifying
Gerais, Brazil.
biofilm formation. The omics analysis may prove to be a promising strategy
E-mail: nagela_bernadelli.mg@hotmail.com
for the diagnosis of biofilms in clinical laboratories since it allows the
2021/1947: received 6 June 2020, revised 1 identification of pathogens in less time than needed for conventional
February 2021 and accepted 23 February techniques and in a more specific manner. However, omic tools are expensive
2021 and require qualified technical expertise, and an analysis of the data obtained
needs to be careful not to neglect subpopulations in the biofilm. More studies
doi:10.1111/jam.15049 must therefore be developed for creating a protocol that guarantees rapid
biofilm identification, ensuring greater chances of success in infection control.
This review discusses the current methods of microbial biofilm detection and
future perspectives for its diagnosis in clinical practice.

symptoms that the patient presents are nonspecific (Jamal


Introduction
et al. 2018). Although there are several methods for diag-
Biofilms are microbial communities adhered to biotic or nosing biofilms in research laboratories, there is no stan-
abiotic surfaces and protected by an extracellular poly- dardized protocol for such a diagnosis in clinical practice.
meric matrix; they have been extensively investigated in Additionally, susceptibility tests to antimicrobials con-
recent decades (Costa-Orlandi et al. 2017). It is estimated tinue to be performed on planktonic bacteria, not repre-
that approximately 65% of microbial infections are senting the clinical reality in the case of an infection
caused by biofilm-forming microorganisms, representing associated with biofilms (Ferrer et al. 2016).
a serious public health problem due to the evasion of the This review discusses the current methods of microbial
host immune system and the resistance of most antimi- biofilm detection and future perspectives for its diagnosis
crobials used for treatment, increasing the persistence of in clinical practice.
the infection (Mooney et al. 2018). Additionally, biofilms
contribute to the increase in morbidity and mortality
Clinical and laboratory diagnosis of microbial
rates and hospital costs, making it difficult to treat and
biofilms
eradicate infections, especially those formed in and on
invasive devices such as catheters and implants (Jamal In 2014, the European Society for Clinical Microbiology
et al. 2018). and Infectious Disease published a guideline to assist
The diagnosis of biofilm-related infections continues to healthcare professionals in the diagnosis of infections
be a challenge since in most infections, the signs and associated with biofilms, with emphasis on clinical and

Journal of Applied Microbiology © 2021 The Society for Applied Microbiology 1


Diagnosis of biofilm infections N.B.S. Silva et al.

laboratory procedures to facilitate diagnosis as well as to (Rajapaksha et al. 2019). To overcome this problem, new
make patient treatment faster and more effective (Høiby and more sophisticated methodologies for the identifica-
et al. 2015). According to this guideline, the most fre- tion of microorganisms in medical devices and clinical
quent clinical signs of an infection associated with bio- samples must be implemented in clinical practice to make
films consist of inflammatory reactions, redness, pain, the identification of microorganisms faster and to facili-
loss of function and fever. The patient must have a his- tate the routine investigation of biofilm formation in
tory of predisposition to infection, such as the use of a clinical practice (Gaudreau et al. 2018; Ordonez et al.
medical device or self-limiting diseases, as well as present- 2019).
ing a persistent infection over more than 7 days and fail- The literature describes several methods of biofilm
ure of antibiotic therapy. Regarding biofilm laboratory detection, and the choice depends on the place of its
diagnosis, the guideline suggests some detection methods, formation and the sample to be analysed; these methods
such as electron microscopy and polymerase chain reac- have greater specificity and sensitivity and are much less
tion (PCR), where it is possible to identify microbial time-consuming than other conventional techniques.
aggregates around inflammatory cells and the presence of Biopsy is considered the most reliable way to detect bio-
mucoid or small cells in positive cultures, indicating films (Ordonez et al. 2019; Rajapaksha et al. 2019).
antibiotic recalcitrance (Kamaruzzaman et al. 2018). Biopsy samples are stained for the visualization of
In the literature, the methods for diagnosis of infec- microbial aggregates, the extracellular matrix of the bio-
tions caused by free-living bacteria, such as those related film and cells of the immune system, and the larger the
to catheter and chronic wounds, are well established size of the tissue and the number of biopsies collected,
(Frickmann et al. 2017; Stoica et al. 2017; Rajapaksha the more sensitive and specific the result will be
et al. 2019). However, when it comes to the diagnosis of (Kamaruzzaman et al. 2018). The viability method of
biofilms, there are few consistent reports and data avail- LIVE/DEAD uses green and red fluorescing markers and
able, especially regarding biofilms formed in and on med- can be used as a qualitative and quantitative technique
ical devices (Høiby et al. 2015). It is estimated that 2% of because it can differentiate healthy cells (stained green)
knee and breast implants, 4% of mechanical heart valves, from damaged cells (stained red) in the biopsy
pacemakers and defibrillators, 10% of ventricular devices (Chevalier et al. 2017).
and 40% of ventricular assist devices are affected by bio- However, in many clinical situations, biopsies cannot
films (Del Pozo 2018). The pathogens are generally be collected or are not indicated, and other samples
derived from the hands of healthcare workers, contami- could be sent to the laboratory, such as sputum, blood,
nated water or fluids and the hospital environment fluids and secretions (Percival 2017). In the laboratory,
(Yadav et al. 2020). Biofilms formed in medical devices analysis of these samples is difficult since the microorgan-
are responsible for the worsening of the patient’s clinical isms in the biofilm are adhered to each other and a sur-
condition. Currently, although many methods allow iden- face, forming a microbial aggregate. Therefore, sonication
tification of biofilms in catheters and other devices, such is generally performed, using energy to agitate particles in
as electronic microscopy, biofilms on these materials are a solution, removing the aggregated and adhered bacteria
not routinely investigated in clinical practice (Stoica et al. from biomaterial surfaces (Galli et al. 2018). After sonica-
2017). tion, the aggregated microorganisms detach themselves
Also, the conventional methods used in clinical labora- from the surface of the material and can be analysed by
tories for the identification of pathogens, which are based several methods, as described below.
on the use of a specific agar, the incubation of the sam-
ple, the isolation of the microorganisms and the enumer-
Polymerase chain reaction
ation of viable cells, can only detect free-living bacteria
and cannot identify biofilm-forming microorganisms, This technique is used for the detection of biofilm-form-
generating results that may not represent the clinical real- ing pathogens directly in clinical samples. It is based on
ity of the patient (Mooney et al. 2018). Besides that, even the amplification of specific regions, providing high
with the use of automated growth detection systems, specificity and sensitivity in the identification of genes
microbial identification takes approximately 5–7 days. In involved in biofilm formation, after the sonication pro-
the wait for the result, mainly with the presence of the cess (Rajapaksha et al. 2019). Some examples of devel-
biofilm, these pathogens have already multiplied, worsen- oped PCR methods are real-time PCR, multiplex PCR
ing the patient’s clinical condition and forcing doctors to and reverse transcriptase PCR. These techniques can be
start empirical therapy even without identifying the used to identify clinical samples that produce biofilms, as
pathogen, which can lead to errors in antibiotic therapy in the study of Shahmoradi et al. (2019), who identified
and, in some cases, to the death of the patient biofilm-forming isolates of Staphylococcus aureus in urine,

2 Journal of Applied Microbiology © 2021 The Society for Applied Microbiology


N.B.S. Silva et al. Diagnosis of biofilm infections

blood, sputum, cerebrospinal fluid, pleural fluid and dyes, which mark the different components of the biofilm
wound samples of hospitalized patients. as well as the extracellular matrix and spatial relationships
Moreover, PCR can be used to identify genes involved of the biofilm (Costa-orlandi et al. 2017).
in biofilm formation, as in the study of Klancnik et al. The analysis of biofilms in vitro contributed to the
(2015), who used the PCR-based method for the identifi- advancement of biofilm research; however, in most exist-
cation of genes involved in the adhesion of different ing methodologies, biofilm is treated and analysed in a
strains of Listeria monocytogenes. different place from where it was formed, which can
damage its structure due to the lack of exact repro-
ducibility of the appropriate temperature, pH and avail-
Fluorescence in situ hybridization
ability of nutrients from the site of origin (Sugimoto
This method consists of the binding of short fluores- et al. 2016). To overcome this problem, a sophisticated
cence-labelled oligonucleotides, which can bind to the microscopic technique can be used in the detection of
specific ribosomal RNA of the target organisms. It has the biofilm in situ, such as multiphoton laser scanning
the advantage of allowing the analysis of the sample with- microscopy, making it possible to analyse the biofilm at
out prior treatment, easily identifying microbial aggre- the site where it was formed, such as catheters and
gates; the samples are analysed via microscopy implants, preserving its structure and architecture. This
(Frickmann et al. 2017). Bernardi et al., (2019) used the methodology is indicated for the acquisition of images of
fluorescence in situ hybridization (FISH) method and autofluorescence materials, where the researcher can
confocal laser scanning microscopy (CLSM) in their obtain high-resolution images of the biofilm structure
study with halitosis to visualize and quantify the biofilm without losing any important components of the biofilm
formed in the dorsum of the tongue, in which they iden- and without previous treatment of the sample (Neu and
tified Fusobacterium nucleatum and Streptococcus sp. bio- Lawrence 2015).
film-forming strains. This method also has been tested in Other types of microscopy are also used in biofilm
bacterial vaginosis, identifying biofilm-forming bacteria in research, such as scanning transmission X-ray micro-
vaginal samples, with greater specificity than for other scopy, which is associated with X-ray absorption spec-
common methods (Machado et al. 2015). troscopy, Hoffman modulation contrast microscopy and
atomic force microscopy; however, they are associated
with high costs (Costa-Orlandi et al. 2017).
Electronic microscopy
Even with the publication of the guidelines proposed
This approach allows the visualization of microbial aggre- by the European Society for Clinical Microbiology and
gates after sonication or FISH. Scanning electron micro- Infectious Disease in 2014 and the current discussion,
scopy (SEM), transmission electron microscopy (TEM) until the present moment, the data about biofilm diagno-
and CLSM are the most used tools in biofilm visualiza- sis and treatment are largely inconsistent, with little
tion (Costa-Orlandi et al. 2017). The high resolution of information to support definitive conclusions for con-
this microscopy allows analysis of all biofilm structures, tainment and prevention strategies. In addition, some
facilitating direct detection and visualization of the bio- proposed methodologies are expensive or require signifi-
film where it was formed, such as in a catheter (Sugi- cant amounts of time and a large number of trained staff,
moto et al. 2016; Schlafer and Meyer 2017). making them impracticable for clinical practice (Mooney
Via SEM, samples can be fixed, dehydrated and et al. 2018).
stained, but in all these processes, mainly in dehydration, However, of all the methodologies mentioned above,
the morphology and structure of the biofilm can PCR and electron microscopy stand out due to their high
be altered, which is a disadvantage of this technique cost-benefit, short experimental time and high sensitivity
(Costa-Orlandi et al. 2017). The TEM is widely used for and specificity in the identification of microorganisms
biofilm architecture visualization and has an advantage (Rajapaksha et al. 2019). These techniques, associated
over SEM since the microbial aggregate is treated with with the evaluation of signs and symptoms and clinical
resin, which ensures greater stability of the extracellular samples of the patient, can modernize and improve the
polymeric matrix of the biofilm. However, it has the dis- way of diagnosing infections in the hospital, generating
advantage of not allowing visualization of the biofilm faster results and using less laboratory inputs than con-
topography (Mohmmed et al. 2017). ventional techniques (Ordonez et al. 2019). In addition,
The CLSM guarantees the 3D visualization of the com- with the use of these methods, the doctor would be able
plete biofilm architecture, besides allowing the identifica- to distinguish infections caused by free-living or sessile
tion of macromolecules such as polysaccharides, proteins, microorganisms, which will reflect in a better prognosis
nucleic acids and lipids through the use of fluorescent for the patient, since the antimicrobial used would be

Journal of Applied Microbiology © 2021 The Society for Applied Microbiology 3


Diagnosis of biofilm infections N.B.S. Silva et al.

more specific and efficient for the infection, decreasing nonforming ones. Furthermore, Mohammadi et al.
the patient’s length of stay as well as the morbidity and (2020) described a significant correlation between phos-
mortality rates in the unit (Høiby et al. 2015). pholipase activity and biofilm production in Candida
albicans isolated from nails, discussing the importance of
biofilm formation to the reduction of azole drug penetra-
Antimicrobial susceptibility testing against
tion into the biofilm matrix and the increased expression
biofilms
of gene resistance associated with antifungal resistance.
Recently, there was a discussion in the literature, on the The relationship between biofilm production and
implementation of a specific breakpoint value to assess antibiotic resistance is not clear; however, a possible
the bacterial susceptibility of biofilm-forming microor- explanation is that the plasmids that harbour antimicro-
ganisms in laboratory diagnosis, but there was low appli- bial resistance genes also harbour genes involved in the
cability; further studies and other pharmacological and production of biofilms (Vuotto et al. 2017), which would
microbiological tests are needed for this value to be used explain why multiresistant microorganisms are also bio-
safely and effectively (Ruiz et al. 2019). Furthermore, it is film producers. In addition, within biofilms, bacteria have
still early to determine a successful antibacterial regimen a cellular communication system called quorum-sensing,
against biofilms, as currently available indirect screening which facilitates the exchange of genes within the com-
tests may generate false negatives in the presence of munity, including antimicrobial resistance genes (Osman
antimicrobial treatment, with the risk of relapse after et al. 2019).
treatment discontinuation (Mooney et al. 2018). Therefore, several bacterial mechanisms may be
Some studies recommend the use of combined antimi- involved in microbial resistance within biofilms, such as
crobial therapy for the treatment of infections associated low antimicrobial penetration due to matrix presence,
with biofilms by Gram-negative bacteria through the use antimicrobial and antifungal release strategies, altered
of macrolides (erythromycin, clarithromycin and azithro- microbial multiplication rate and bacterial physiological
mycin); however, determining a specific antibiotic ther- changes (Singh et al. 2017).
apy for these infections is difficult due to the variety of In 1999, the first strategy of the minimum biofilm
microorganisms that live in biofilms (Cepas et al. 2019). inhibitory concentration (MBIC) and the minimum bio-
This makes it difficult to determine a minimum inhibi- film eradication concentration (MBEC) was created using
tory concentration (MIC) of eradication for these sessile the Calgary biofilm device method. In this technique,
microorganisms since to inhibit a biofilm, a drug at a biofilm formation is tested with stakes that fit into the
high concentration is required, and considering the side wells of the microtiter plate, containing growth medium
effects and toxicity that antimicrobials have on human and bacteria. Thus, microbes do not sediment and only
cells, this type of treatment becomes impracticable develop in the sessile form (Azeredo et al. 2017). Since
(Mooney et al. 2018). then, other procedures have been proposed, such as mod-
Bacteria and fungi in biofilms are more resistant to els based on bioreactors, flow cell systems and MBIC def-
antimicrobials than free-living microorganisms, with min- initions evaluating biomass, but the application and
imum eradication concentrations of 10–1000 times higher standardization of these procedures in the clinic are still
compared to planktonic bacteria (Mooney et al. 2018). In pending (Di Luca et al. 2017).
biofilms, gene expression is altered, with negative regula- The MBIC of biofilm and the MBEC are used to
tion of genes associated with glycolysis, carbon metabo- determine the efficacy of antimicrobials against biofilms
lism and ribosomal proteins. In addition, the (Azeredo et al. 2017). These values are generally consider-
transcription of genes encoding peptideoglycan, cell wall ably higher than the MIC of planktonic bacteria. Some
synthesis and efflux pump activity are greater in sessile studies have been successful in developing drugs for the
cells than in planktonic cells (Castro et al. 2017; Nielsen treatment of catheter-related infections, taking into
et al. 2019). account the MBIC and MBEC values of the biofilm, while
Vuotto et al. (2017), in their study with Klebsiella other clinical trials have failed to replicate these results
pneumoniae urinary strains, demonstrated a correlation with other microorganisms from different sites. In addi-
between biofilm production and resistance profile, show- tion, most microbial resistance studies of biofilms are
ing that the samples that showed extensive resistance to based on in vitro assays, questioning whether these tests
antimicrobials showed a greater ability to form biofilms can be reproduced in vivo (Coenye et al. 2018).
compared with sensitive samples. Neopane et al. (2018) Although several microbial susceptibility tests on bio-
also observed that the S. aureus biofilm-forming samples films have already been described, so far, none has been
collected from wounds of hospitalized patients showed approved as a reference method in clinical microbiology,
higher levels of microbial resistance when compared to generating an increasing interest in the development of

4 Journal of Applied Microbiology © 2021 The Society for Applied Microbiology


N.B.S. Silva et al. Diagnosis of biofilm infections

new specific sensitivity tests for sessile bacteria (Singh and red indicating no biofilm production (Lee et al.
et al. 2017). 2015). This is a quantitative test because it is based on a
subjective chromatic evaluation (Melo et al. 2013). Solati
et al. (2015) have used it to assess Staphylococcus epider-
Advances in scientific research: In vitro and
midis produced by biofilm collected from the urine and
in vivo biofilm detection methods
blood of patients with urinary tract infections and sep-
Many in vitro and in vivo methods have been used for a ticemia, respectively, in which they identified 55% of the
better understanding of the biology of biofilms and their isolates with the greatest potential for biofilm formation;
detection. Colorimetric assays are the most widely used most were collected from urine.
ones and commonly applied in studies related to the
development and susceptibility of biofilms to drugs and Tube biofilm formation test
the quantification of specific biofilm structures (Shukla This test consists of the use of plastic tubes containing
and Rao 2017). These methods, although requiring signif- sample inoculum stained with 01% violet crystal. After
icant amounts of time and a high number of trained several washing processes with phosphate buffer saline
staff, are important allies in identifying biofilm formation and fixation with sodium acetate, the tubes are inverted
and for a better understanding, mainly of the susceptibil- in an adsorbent paper, and those with the presence of the
ity of biofilms to anti-biofilms and antimicrobial com- stained film on the bottom and walls of the tube, indicat-
pounds, which can assist in the development of new ing the presence of biofilm, are being considered positive
methods of diagnosis and treatment that can be useful in (Halim et al. 2018).
clinical practice (Qu et al. 2017). The main in vitro and Some studies have used this method to detect clinical
in vivo tests used by the academic community are listed isolates producing biofilms; for example, Neopane et al.
in Table 1 and described below. (2018) have identified 30 S. aureus biofilm-forming iso-
lates collected from wounds of hospital-admitted patients,
while Hassan and Khider (2019) have evaluated the cor-
In vitro biofilm detection models
relation of the formation of biofilms in clinical samples
Congo red agar test of Acinetobacter baumannii using the tube biofilm forma-
This agar allows the differentiation of the colonies in two tion test and molecular tests. Despite being a low-cost
colours, with black indicating strong biofilm production test, it is a qualitative and subjective test.

Table 1 Main in vitro and in vivo biofilm detection methods

Method Advantages Disadvantages References

In vitro biofilm detection


Congo red agar High accuracy in the detection of Low reproducibility Melo et al. (2013)
test Staphylococcus sp. biofilm producers
Tube biofilm Low cost Subjective reading Halim et al. (2018)
formation test
Microplate test Low cost Lack of standardization in the interpretation of Qu et al. (2017)
Several tests can be done simultaneously results
Crystal violet Low cost Low specificity Xu et al. (2016)
Simple technique
High replicability
Safranin Non-toxic dye Low replicability and sensitivity Stepanovıc et al. (2007);
Ommen et al. (2017)
XTT Simple technique High cost Costa-Orlandi et al.
High replicability Low sensitivity salt Retention by the pathogens (2017)
that can interfere with the result
In vivo biofilm detection
D. melanogaster High homologies between the Drosophila Preference for yeasts and Escherichia coli for Yamaguchi and Yoshida
and human genomes having small genomes (2018)
Easy to handle Does not have haemoglobin
Inexpensive to maintain
C. elegans Powerful methods for studying physiological Nematode culture standardization factors may Park et al. (2017)
processes interfere with its survival

Journal of Applied Microbiology © 2021 The Society for Applied Microbiology 5


Diagnosis of biofilm infections N.B.S. Silva et al.

component, which is replaced by DNA and other com-


Microplate test
pounds. In addition, each microorganism has a type of
This test uses microplates with flat bottom or U-shaped
extracellular matrix which varies in its cellular localisa-
wells, where all stages of biofilm formation occur within
tion, chemical composition, nature and functions; many
these wells (Di Luca et al. 2017). It is a cheap technique
are polyanionic, but others are neutral or polycationic
that requires few reagents, and several tests can be done
(Limoli et al. 2015).
simultaneously. For the detection of biofilm production,
the wells with the samples are stained with specific dyes
that quantify the main structures of the biofilm. This part
The use of 2,3-bis (2-methoxy-4-nitro-5-sulfophenyl)-5-
of the experiment is the so-called ‘colorimetric test’ (Qu
[(phenylamino) carbonyl 2H-tetrazolium hydroxide
et al. 2017). According to the absorbance result of each
(XTT). The tetrazolic salt XTT is used to evaluate the
isolate in these colorimetric tests, it is possible to classify
number of viable cells and the metabolic activity of bio-
them into nonproducers, weak, moderate or strong pro-
films. When microorganisms in the biofilm produce
ducers of biofilms. However, this classification is highly
metabolites, XTT changes their chemical conformation
subjective and can vary according to the species and the
and is transformed into formazan, which is seen in the
cut-off value that the researcher uses as a reference
microtiter plate by a colour change from neutral to
(Shukla and Rao 2017).
orange (Costa-Orlandi et al. 2017). This compound is
most frequently used in detecting and evaluating the
metabolic activity of biofilms produced by fungi, as in
Crystal violet. In addition to the use of this dye in tube the study by Di Domenico et al. (2018), who detected 38
tests, it can also be applied in microplate tests for an clinical isolates of Candida sp. biofilm producers, but it
assessment of the total biomass of the biofilm. It is a can also be used for bacterial evaluation, as in the study
cost-efficient substance, and the test is easy to interpret by Xu et al. (2016), who reported that 77% of S. aureus
and has high replicability (Shukla and Rao 2017). How- isolates were biofilm-forming.
ever, this dye has the disadvantage of staining the entire Some researchers use (3-[4,5-dimethylthiazol-2-yl]-
structure of the biofilm, without specificity. Moreover, 2,5iphenyltetrazolium bromide) MTT, which is a salt that
after being deposited in the plate, the wells need to be behaves in a similar way to XTT, albeit with a colour
washed, culminating in the loss of important biofilm change from neutral to purple. Comparing the two salts,
components and structures (Xu et al. 2016). It is the XTT has more advantages because after its reduction in
most used colorimetric test in the microplate test, making formazan, the wells can be read immediately, while MTT
it possible to quantify the entire biomass and matrix of requires other steps prior to reading, such as cell lysis
biofilms (Di luca et al. 2017). (Costa-Orlandi et al. 2017). Another disadvantage of the
In the literature, there are several studies that use crys- MTT salt is the insolubility of formazan in water, making
tal violet in the identification and quantification of bio- it necessary to dissolve the precipitate before measuring
film production in bacteria and fungi (Xu et al. 2016; absorbance. In addition, reduced MTT can react with
Vuotto et al. 2017; Mohammadi et al. 2020). some compounds, forming large intracellular deposits,
capable of damaging microbial cells during the test, pro-
ducing false positive or negative results (Grela et al.
Safranin. Safranin is a nontoxic dye, and its use is simi- 2015).
lar to that of crystal violet. However, it is less frequently However, XTT has some disadvantages, such as the
used in the quantification of biomass and results in lower lack of a linear relationship between the number of
optical densities than crystal violet staining (Ommen microorganisms present in the biofilm and the colorimet-
et al. 2017). Some authors, such as Babapour et al. ric signal, salt retention by these pathogens in planktonic
(2016) and Karami et al. (2020), have used safranin as a and sessile form and a high cost (Alonso et al. 2017).
dye in the microplate test for the identification of clinical Additionally, XTT can react with some nanoparticles and
isolates of biofilm producers. also be reduced by O2. Toxicity testing of engineered
Some researchers believe that this dye specifically stains nanomaterials including nano-TiO2 has generated a many
the mucopolysaccharides of the extracellular matrix number of publications in the last several years (Wang
because of their high affinity with these compounds. et al. 2011). Furthermore, problems regarding intraspecies
Mucopolysaccharides are molecules produced by cells and interspecies variability have been reported, making
within the biofilm that will compose the extracellular the use of these methods unfeasible in some susceptibility
matrix of the biofilm (Stepanovic et al. 2007). However, tests for both planktonic cells and biofilms (Peeters et al.
not all biofilms have polysaccharides as a matrix 2008).

6 Journal of Applied Microbiology © 2021 The Society for Applied Microbiology


N.B.S. Silva et al. Diagnosis of biofilm infections

problem encountered by researchers is that the same


Detection of biofilms in vivo
microorganisms may present different mass spectra when
Some researchers have used animal models to advance analysed repeatedly (Singhal et al. 2015). Another limita-
the research and detection of biofilms in vivo. Due to tion of the technique is the difficulty in differentiating
some difficulties encountered by the researchers in mam- microbial samples with a high degree of similarity due to
malian studies, including the approval of the Ethics Com- the several similar compounds included in their cells. In
mittee for such studies, other nonmammalian models are addition, other intracellular compounds that vary within
being used, such as Drosophila melanogaster (McLaughlin each microorganism may not be detected by the equip-
and Bratu 2015) and Caenorhabditis elegans (Park et al. ment; especially if they belong to low- or high-mass pro-
2017). teins, which are generally not ionized by the apparatus
For example, Kamareddine et al. (2018) have used (Mlynarikova et al. 2016). However, according to Gau-
D. melanogaster in their study of infection by Vibrio cho- dreau et al. (2018), sample preparation for analysis is cru-
lerae, where they discovered that quorum-sensing, besides cial for the success of the test, and when the sample is
being a cellular communication mechanism in biofilms, transferred directly to the analysis, together with the pres-
also exercises control over the host’s metabolic pathways. ence of formic acid and the matrix, the chances of inter-
Another important study model for biofilms is C. elegans, ference are lower compared to other types of preparation
which is used for the evaluation of the progression of with acetonitrile, water and ethanol.
intestinal infection caused by Salmonella sp., where it was New studies are necessary to prove the efficacy of this
possible to identify a toxin that is secreted by these bacte- methodology in the detection of biofilms, since it pre-
ria in biofilm formation (Desai et al. 2019). sents limitations and low reproducibility. While MALDI-
The advantages of such an approach are the ease of TOF is not yet fully effective in the detection of biofilms,
handling and the lower costs. Moreover, the small size of conventional in vitro and in vivo techniques are still most
the models makes it easy to keep them in microtiter frequently used in research labs (Singhal et al. 2015).
plates, thus facilitating the high-performance screening of
biofilm formation and making them important tools in
Omics analysis: The role of proteomic,
understanding the pathogen-host relationship (Costa-
transcriptomic and metabolomic analyses in the
Orlandi et al. 2017).
diagnosis of biofilms
The traditional methods of microbiological diagnosis in
Matrix-assisted laser desorption ionization time-
clinical laboratories include the growth and isolation of
of-flight mass spectrometry (MALDI-TOF MS): Is it
the microorganism, the detection of antibodies or anti-
the diagnostic method of the future?
gens in serological tests and, in some laboratories, genetic
In recent years, mass spectrophotometry has been used identification (DNA or RNA) through the PCR technique
by some clinical laboratories for the identification of bac- (Ordonez et al. 2019). While most molecular tests target
teria and fungi. It is a method that compares the micro- only a limited number of pathogens using specific pri-
bial protein profile, obtained from a database of reference mers, omics analysis allows to analyse the entire micro-
mass spectra, for the identification of genera, species and biome of the pathogen and to identify specific proteins
subspecies. Microbial identification is performed in 60 s and metabolites produced by pathogens (Otto et al.
and has been used in the diagnosis of several infectious 2012). These technologies are also being developed to
diseases around the world (Singhal et al. 2015). Cur- detect differences in the transcription, translation and
rently, this methodology is the target of researchers and metabolism of microorganisms in biofilms, describing
applied biofilm studies, since MALDI-TOF allows the dif- important information of these communities, such as the
ferentiation of planktonic cells and biofilms due to pro- influence of environmental factors on biofilm adhesion
files of different mass spectra (Caputo et al. 2018). The and formation as well as physiological and mutational
method was tested on different species of Gram-positive factors (Chiu and Miller 2019). Table 2 shows the main
and negative bacteria and fungi (Kubesova et al. 2012; methods used in the omics analysis as well as the samples
Pereira et al. 2015; Caputo et al. 2018), showing that it is that can be evaluated and the infections that can be diag-
possible, through MALDI-TOF, to differentiate microor- nosed.
ganisms that form weak or strong biofilms, based on The study of proteomics in biofilms allows the identifi-
their molecular mass size. cation of specific proteins that are produced in this com-
However, due to several problems and limitations of munity, as well as the distribution, posttranslational
this technique, MALDI-TOF is far from being the gold modifications, structure, function and quantity of these
standard for identifying biofilm formation. A common proteins (Washio and Takahashi 2016). The success of

Journal of Applied Microbiology © 2021 The Society for Applied Microbiology 7


Diagnosis of biofilm infections N.B.S. Silva et al.

Table 2 Omics analysis in the clinical diagnosis of biofilms

Detection methods Clinical samples Clinical indication Reference

Transcriptomic
The express sequence tag (EST) Blood Sepsis Hrdlickova et al. (2017)
Serial Gene Expression Analysis (SAGE) Peripheral blood mononuclear cel Urinary Tract Infection
DNA microarrays Several body fluids and tissues Multiplexed pathogen detection
RNA-Seq (4sU-Seq, RIP-Seq, (Riboseq)) Cerebrospinal fluid Meningitis
Proteomic
2D gels (DIGE) Blood Sepsis Otto et al. (2012)
Gel free (iTRAQ, TMT)
Spiked-in peptides Secretions Urinary Tract Infection
Label-free quantitation Cell-free body fluids Respiratory infections
Metabolomic
Capillary electrophoresis (CE) Blood Sepsis Chiu and Miller (2019)
Fluids Washio and Takahashi
(2016)
Time-of-flight mass spectrometer Stool sample Infections by Clostridium difficile
(TOF-MS)
CE-TOFMS Cerebrospinal fluid Meningitis

proteomics studies is due to the invention of the 2DE molecules on a large scale, such as the express sequence
gel, which is applied in orthogonal separation to classify tag (EST) method, serial gene expression analysis (SAGE)
species according to their molecular weight, where sam- and DNA microarrays; however, they have numerous dis-
ples are stained and identified using software (Otto et al. advantages, such as high costs, and demand for informa-
2012). Despite being a technique similar to MALDI-TOF, tion on the sample genome and transcriptome, impeding
it is more sensitive and can differentiate nonstressed from discoveries (Hrdlickova et al. 2017). The method of
stressed cells, cellular mutations and different time points choice for studying gene expression and identifying new
from the same organism. The combination of proteomics species of RNA is RNA-Seq, as it offers less background
analysis with the use of 2D gel and MALDI-TOF can gen- noise and greater detection than previous methodologies
erate more sensitive and specific data, revealing cases of (Hrdlickova et al. 2017). The transcriptomic approach is
multiple proteins found in the same place and those with of great interest in biofilm research, as it allows the detec-
low mass, reducing false values and improving the disad- tion of specific genes in the formation of biofilms and
vantages of MALDI-TOF (Otto et al. 2012; Singhal et al. new mechanisms of survival and antimicrobial resistance
2015). (Chiu and Miller 2019). For example, Kean et al. (2018)
Erdmann et al. (2019) used proteomics analysis and used this approach to investigate the mechanisms of bio-
identified 1021 different proteins in the biofilms pro- film formation and resistance in Candida auris, a recently
duced by 27 samples of Pseudomonas aeruginosa and a discovered yeast.
protein for mRNA that was common among species, The metabolic analysis is based on the identification
even under different conditions, suggesting a posttran- and quantification of metabolite features in living cells,
scriptional pattern. If researchers were able to identify including gene transcription, mRNA translation, protein
proteins common in most biofilms produced, it would analysis and enzymatic reactions (Zhang and Powers
be possible, through the methods mentioned above, to 2012). In the 2000s, several devices with a metabolic
reduce the time needed for the identification of approach were used, such as capillary electrophoresis and
microorganisms, facilitating our understanding of the time-of-flight mass spectrometer (TOF MS), which iden-
main regulatory pathways and adaptation strategies for tify metabolites according to their ionic charge and mass
bacterial biofilms, which can be useful in the develop- spectrometry, respectively. By connecting these two
ment of new treatment strategies for these biofilms devices, these techniques can separate small ionic mole-
(Chiu and Miller 2019). cules and accurately measure their masses; they have been
Transcriptomics analysis allows identifying an RNA widely used since then. The metabolic approach provides
molecule in a live cell. In the 1990s, several methodolo- data that allow the diagnosis of a disease as well as its
gies were created that allowed sequencing specific RNA progress (Washio and Takahashi 2016).

8 Journal of Applied Microbiology © 2021 The Society for Applied Microbiology


N.B.S. Silva et al. Diagnosis of biofilm infections

This allows, for example, the identification of chemical is needed to perform molecular analyses (Azeredo et al.
signalling molecules, so-called ‘autoinducers’, responsible 2017).
for cellular communication within biofilms. Autoinducers Another question that remains under investigation con-
are targets of promising drugs for biofilms since these cerns the reference to compare the omic data obtained.
molecules are not present in the human organism and Often a planktonic cell is used as a control, but it may not
can therefore be used as biofilm markers (Mooney et al. be the most appropriate, since the biofilm has several
2018). The molecular structure of a biofilm can positively stages of development and any attempt at correlation
impact the biology of systems and, consequently, the would be incorrect. The best choice for the investigation
development of new diagnostic and/or therapeutic tools of biofilm physiology remains to compare a wild-type
(Zhang and Powers 2012). strain with an isogenic mutant (Zhang and Powers 2012).
The biofilm matrix can also be a problem for proteomic
analysis since it can make access to different intracellular
Advantages and disadvantages of omic analysis
membrane and parietal subproteomes difficult (Washio
The new omic approaches allow an evaluation of several and Takahashi 2016). Also, the analysis of exoproteoma,
biological units of biofilms, such as gene expression, pro- that is, proteins that are secreted into the extracellular
teins and metabolites. This generates highly relevant data environment, is also a challenge (Desvaux et al. 2009). In
for a better understanding of biofilms, allowing the dis- addition, omic analysis requires qualified technical exper-
covery of new biomarkers for various diseases and the tise and is expensive.
development of new methods of diagnostics and therapies Despite the challenges that these techniques still con-
(Chiu and Miller 2019). The application of these omic front, the development of omic technologies is essential
methods in the health area can generate opportunities for for a better understanding of the development and evolu-
clinical interventions, as in the study by Nascimento tion of biofilms, allowing the creation of new methods of
et al. (2017), who through omic analyses identified new diagnosis and treatments in the health area (Chiu and
oral Streptococcus and their phenotypic characterization Miller 2019).
revealed promising candidates for probiotic therapy. To
investigate complex multispecies biofilms, the use of omic
Conclusions
tools is the promise of a breakthrough regarding the
understanding of bacterial interaction and physiology Despite all advances in research, biofilm-producing
within biofilms. microorganisms remain a serious public health problem,
However, although the transcriptomic, proteomic and often neglected in various health areas. Biofilm con-
metabolomic analysis are complementary methods to tributes to increased microbial resistance, making treat-
investigate the cellular physiology of biofilms, these omic ment more difficult and worsening the clinical conditions
methods were not designed to be evaluated together; that of patients, contributing to the increase in morbidity and
is, it is not possible to validate or compare one data with mortality rates. The diagnosis of biofilms is challenging,
the other (Azeredo et al. 2017). Also, biofilm is by defini- since the current screening methodologies performed in
tion heterogeneous, varying according to its location, the clinic, such as susceptibility tests, detect only plank-
stage of development and physiology of microbial cells. tonic colonies, not representing the clinical reality of the
In the omic analysis, the data result from a cell that will patient.
represent the entire population, consequently, a subpopu- Also, even with several biofilm detection methods
lation can be neglected or be different from the cell col- described in the literature, there is currently no routine
lected for analysis. test or protocol established for this purpose in the clinic.
Therefore, the results of the omic analysis should be Omics analysis is a promising form of diagnosis in clini-
analysed with caution, considering that the results cal laboratories since it allows the identification of patho-
obtained are averages with margins of error (Jamal gens in less time than needed for conventional
et al. 2018). However, flow cell cameras can work techniques, and it is possible to analyse the entire patho-
around this problem, as microbial cells grow statically. gen microbiome, including an identification of produc-
Another solution is the laser capture microdissection tion of proteins and metabolites, which allows a better
microscopy as it allows the comparison and identifica- understanding of its pathogenesis and a more specific
tion of the physiology of cells and their special distribu- identification.
tion within the biofilm, for example, the differences More studies must therefore be developed to facilitate
between cells that reside on the periphery of biofilms the development of efficient and reliable protocols for
with those that live in the superficial regions. The main rapid biofilm identification, ensuring greater chances of
challenge that a remains unsolved is how much biomass success in infection control in clinical practice.

Journal of Applied Microbiology © 2021 The Society for Applied Microbiology 9


Diagnosis of biofilm infections N.B.S. Silva et al.

Conflicts of Interest Melo, W.C.M.A. et al. (2017) Fungal biofilms and


polymicrobial diseases. J Fungi 3, 22. https://doi.org/10.
No conflicts of interest are declared. 3390/jof3020022.
Del Pozo, J.L. (2018) Biofilm-related disease. Expert Rev Anti
References Infect Ther 16, 51–65. https://doi.org/10.1080/14787210.
2018.1417036.
Alonso, B., Cruces, R., Perez, A., Sanchez-Carrillo, C. and Desai, S.K., Padmanabhan, A., Harshe, S., Zaidel-Bar, R. and
Guembe, M. (2017) Comparison of the XTT and resazurin Kenney, L.J. (2019) Salmonella biofilms program innate
assays for quantification of the metabolic activity of immunity for persistence in Caenorhabditis elegans. Proc
Staphylococcus aureus biofilm. J Microbiol Methods 139, Natl Acad Sci 116, 12462–12467. https://doi.org/10.1073/
135–137. https://doi.org/10.1016/j.mimet.2017.06.004. pnas.1822018116.
Azeredo, J., Azevedo, N.F., Briandet, R., Cerca, N., Coenye, T., Desvaux, M., Hebraud, M., Talon, R. and Henderson, I.R.
Costa, A.R., Desvaux, M., Bonaventura, G.D. et al. (2017) (2009) Secretion and subcellular localizations of bacterial
Critical review on biofilm methods. Crit Ver Microbiol 43, proteins: a semantic awareness issue. Trends Microbiol 17,
313–351. https://doi.org/10.1080/1040841X.2016.1208146. 139–145. https://doi.org/10.1016/j.tim.2009.01.004.
Babapour, E., Haddadi, A., Mirnejad, R., Angaji, S.A. and Di Domenico, E.G., Cavallo, I., Guembe, M., Prignano, G.,
Amirmozafari, N. (2016) Biofilm formation in clinical Gallo, M.T., Bordignon, V., D’Agosto, B., Sperduti, I.
isolates of nosocomial Acinetobacter baumannii and its et al. (2018) The clinical Biofilm Ring Test: a promising
relationship with multidrug resistance. Asian Pac J Trop tool for the clinical assessment of biofilm-producing
Biomed 6, 528–533. https://doi.org/10.1016/j.apjtb.2016.04. Candida species. FEMS Yeast Res 18. https://doi.org/10.
006. 1093/femsyr/foy025.
Bernardi, S., Continenza, M.A., Al-Ahmad, A., Karygianni, L., Di Luca, M., Navari, E., Esin, S., Menichini, M., Barnini, S.,
Follo, M., Filippi, A. and Macchiarelli, G. (2019) Trampuz, A., Casani, A. & Batoni, G. (2017) Detection of
Streptococcus spp. and Fusobacterium nucleatum in tongue biofilms in biopsies from chronic rhinosinusitis patients:
dorsum biofilm from halitosis patients: a fluorescence in vitro biofilm forming ability and antimicrobial
in situ hybridization (FISH) and confocal laser scanning susceptibility testing in biofilm mode of growth of isolated
microscopy (CLSM) study. New Microbiol 42, 108–113. bacteria. In Donelli, G., ed. Advances in Microbiology,
Caputo, P., Di Martino, M.C., Perfetto, B., Iovino, F. and Infectious Diseases and Public Health, Vol. 1057, pp. 127.
Donnarumma, G. (2018) Use of MALDI-TOF MS to Advances in Experimental Medicine and Biology. Cham:
discriminate between biofilm-producer and non-producer Springer. https://doi.org/10.1007/5584_2017_34
strains of Staphylococcus epidermidis. Int J Environ Res Erdmann, J., Th€ oming, J.G., Pohl, S., Pich, A., Lenz, C. and
Public Health 15, 1695. https://doi.org/10.3390/ije H€aussler, S. (2019) The core proteome of biofilm-grown
rph15081695. clinical Pseudomonas aeruginosa isolates. Cells 8, 1129.
Castro, J., Franca, A., Bradwell, K.R., Serrano, M.G., Jefferson, https://doi.org/10.3390/cells8101129.
K.K. and Cerca, N. (2017) Comparative transcriptomic 
Ferrer, M.D., Rodriguez, J.C., Alvarez, L., Artacho, A., Royo,
analysis of Gardnerella vaginalis biofilms vs. planktonic G. and Mira, A. (2016) Effect of antibiotics on biofilm
cultures using RNA-seq. NPJ Biofilms Microbiomes 3, 1–7. inhibition and induction measured by real-time cell
Cepas, V., Lopez, Y., Mu~ noz, E., Rolo, D., Ardanuy, C., Martı, analysis. J Appl Microbiol 122, 640–650. https://doi.org/10.
S., Xercavins, M., Horcajada, J.P. et al. (2019) 1111/jam.13368.
Relationship between biofilm formation and antimicrobial Frickmann, H., Zautner, A.E., Moter, A., Kikhney, J., Hagen,
resistance in Gram-negative bacteria. Microbial Drug Resist R.M., Stender, H. and Poppert, S. (2017) Fluorescence
25, 72–79. https://doi.org/10.1089/mdr.2018.0027. in situ hybridization (FISH) in the microbiological
Chevalier, M., Ranque, S. and Pr^echeur, I. (2017) Oral fungal- diagnostic routine laboratory: a review. Crit Rev Microbiol
bacterial biofilm models in vitro: a review. Med Mycol 56, 43, 263–293. https://doi.org/10.3109/1040841X.2016.
653–667. https://doi.org/10.1093/mmy/myx111. 1169990.
Chiu, C.Y. and Miller, S.A. (2019) Clinical metagenomics. Nat Galli, J., Calo, L., Meucci, D., Giuliani, M., Lucidi, D., Paludetti,
Rev Genet 20, 341. https://doi.org/10.1038/s41576-019- G., Torelli, M.S., Sanguinetti, M. et al. (2018) Biofilm in
0113-7. voice prosthesis: a prospective cohort study and laboratory
Coenye, T., Goeres, D., Bambeke, F.V. and Bjarnsholt, T. tests using sonication and SEM analysis. Clin Otolaryngol
(2018) Should standardized susceptibility testing for 43, 1260–1265. https://doi.org/10.1111/coa.13141.
microbial biofilms be introduced in clinical practice? Clin Gaudreau, A.M., Labrie, J., Goetz, C., Dufour, S. and Jacques,
Microbiol Infect 24, 570–572. https://doi.org/10.1016/j.cmi. M. (2018) Evaluation of MALDI-TOF mass spectrometry
2018.01.003. for the identification of bacteria growing as biofilms. J
Costa-Orlandi, C.B., Sardi, J.C.O., Pitangui, N.S., Oliveira, Microbiol Methods 145, 9–81. https://doi.org/10.1016/j.
H.C., Scorzoni, L., Galeane, M.C., Medina-Alarc on, K.P., mimet.2018.01.003.

10 Journal of Applied Microbiology © 2021 The Society for Applied Microbiology


N.B.S. Silva et al. Diagnosis of biofilm infections

Grela, E., Zaz bek, A. and Grabowiecka, A. (2015) Interferences in techniques and MALDI-TOF mass spectrometry for
the optimization of the MTT assay for viability estimation phenotypical discrimination. Analyst 137, 1937–1943.
of Proteus mirabilis. Avicenna J Med Biotechnol 7, 159. Lee, J.S., Bae, Y.M., Lee, S.Y. and Lee, S.Y. (2015) Biofilm
Halim, R.M.A., Kassem, N.N. and Mahmoud, B.S. (2018) formation of Staphylococcus aureus on various surfaces and
Detection of biofilm producing staphylococci among their resistance to chlorine sanitizer. J Food Sci 80, M2279.
different clinical isolates and its relation to methicillin https://doi.org/10.1111/1750-3841.13017.
susceptibility. Open Access Maced J Med Sci 6, 1335. Limoli, D.H., Jones, C.J. and Wozniak, D.J. (2015) Bacterial
https://doi.org/10.3889/oamjms.2018.246. extracellular polysaccharides in biofilm formation and
Hassan, P.A. and Khider, A.K. (2019) Correlation of biofilm function. Microbial Biofilms 223–247, https://doi.org/10.
formation and antibiotic resistance among clinical and soil 1128/9781555817466.ch11.
isolates of Acinetobacter baumannii in Iraq. Acta Microbiol Machado, A., Castro, J., Cereija, T., Almeida, C. and Cerca, N.
et Immunol Hungarica 1–10. https://doi.org/10.1556/030. (2015) Diagnosis of bacterial vaginosis by a new multiplex
66.2019.026. peptide nucleic acid fluorescence in situ hybridization
Høiby, N., Bjarnsholt, T., Moser, C., Bassi, G.L., Coenye, T., method. PeerJ 3, e780. https://doi.org/10.7717/peerj.780.
Donelli, G., Hall-Stoodley, L., Imbert, C. et al. (2015) McLaughlin, J.M. and Bratu, D.P. (2015) Drosophila
ESCMID guideline for the diagnosis and treatment of melanogaster oogenesis: an overview. In Bratu, D. and
biofilm infections 2014. Clin Microbiol Infect 21, S1–S25. McNeil, G., eds. Drosophila Oogenesis, pp 1–20. New York,
https://doi.org/10.1016/j.cmi.2014.10.024. NY: Humana Press.
Hrdlickova, R., Toloue, M. and Tian, B. (2017) RNA-Seq Melo, P.C., Ferreira, L.M., Filho, A.N., Zafalon, L.F., Vicente,
methods for transcriptome analysis. Adv Rev 8, e1364. H.I.G. and Souza, V. (2013) Comparison of methods for
https://doi.org/10.1002/wrna.1364. the detection of biofilm formation by Staphylococcus
Jamal, M., Ahmad, W., Andleeb, S., Jalil, F., Imran, M., aureus isolated from bovine subclinical mastitis. Braz J
Nawaz, M.S., Hussain, T., Ali, M. et al. (2018) Bacterial Microbiol 44, 119–124.
biofilm and associated infections. J Chin Med Assoc 81, Mlynarikova, K., Sedo, O., R
uzicka, F., Zdrahal, Z., Hola, V.
1–5. https://doi.org/10.1016/j.jcma.2017.07.012. and Mahelova, M. (2016) Evaluation of capacity to detect
Kamareddine, L., Wong, A.C.N., Vanhove, A.S., Hang, S., ability to form biofilm in Candida parapsilosis sensu
Purdy, A.E., Kierek-Pearson, K., Asara, J.M., Ali, A. Jr stricto strains by MALDI-TOF MS. Folia Microbiol 61,
et al. (2018) Activation of Vibrio cholerae quorum sensing 465–471. https://doi.org/10.1007/s12223-016-0458-7.
promotes survival of an arthropod host. Nat Microbiol 3, Mohammadi, F., Ghasemi, Z., Familsatarian, B., Salehi, E.,
243–252. Sharifynia, S., Barikani, A., Mirzadeh, M. and Hosseini,
Kamaruzzaman, N.F., Tan, L.P., Yazid, K.A.M., Saeed, S.I., M.A. (2020) Relationship between antifungal susceptibility
Hamdan, R.H., Choong, S.S., Wong, W.K., Chivu, A. profile and virulence factors in Candida albicans isolated
et al. (2018) Targeting the bacterial protective armour; from nail specimens. Rev Soc Bras de Med Trop 53,
challenges and novel strategies in the treatment of https://doi.org/10.1590/0037-8682-0214-2019.
microbial biofilm. Materials 11, 1705. https://doi.org/10. Mohmmed, S.A., Vianna, M.E., Penny, M.R., Hilton, S.T.,
3390/ma11091705. Mordan, N. and Knowles, J.C. (2017) Confocal laser
Karami, P., Khaledi, A., Mashoof, R.Y., Yaghoobi, M.H., scanning, scanning electron, and transmission electron
Karami, M., Dastan, D. and Alikhani, M.Y. (2020) The microscopy investigation of Enterococcus faecalis biofilm
correlation between biofilm formation capability and degradation using passive and active sodium hypochlorite
antibiotic resistance pattern in Pseudomonas aeruginosa. irrigation within a simulated root canal model.
Gene Reports 18, https://doi.org/10.1016/j.genrep.2019. Microbiology Open 6, e00455. https://doi.org/10.1002/mb
100561. o3.455.
Kean, R., Delaney, C., Sherry, L., Borman, A., Johnson, E.M., Mooney, J.A., Pridgen, E.M., Manasherob, R., Suh, G.,
Richardson, M.D., Rautemaa-Richardson, R., Williams, C. Blackwell, H.E., Barron, A.E., Bollyky, P.L., Goodman,
et al. (2018) Transcriptome assembly and profiling of S.G. et al. (2018) Periprosthetic bacterial biofilm and
Candida auris reveals novel insights into biofilm-mediated quorum sensing. J Orthop Res 36, 2331–2339. https://doi.
resistance. Msphere 3, https://doi.org/10.1128/mSphere. org/10.1002/jor.24019.
00334-18. Nascimento, M.M., Zaura, E., Mira, A., Takahashi, N. and Ten
Klancnik, A., Toplak, N., Kovac, M., Marquis, H. and Jersek, Cate, J.M. (2017) Second era of OMICS in caries research:
B. (2015) Quantification of Listeria monocytogenes cells moving past the phase of disillusionment. J Dent Res 96,
with digital PCR and their biofilm cells with real-time 733–740. https://doi.org/10.1177/0022034517701902.
PCR. J Microbiol Methods 118, 37–41. https://doi.org/10. Neopane, P., Nepal, H.P., Shrestha, R., Uehara, O. and
1016/j.mimet.2015.08.012. Abiko, Y. (2018) In vitro biofilm formation by
Kubesova, A., Salplachta, J., Horka, M., Ruzicka, F. and Slais, Staphylococcus aureus isolated from wounds of hospital-
K. (2012) Candida ´´Psilosis´´ - electromigration admitted patients and their association with antimicrobial

Journal of Applied Microbiology © 2021 The Society for Applied Microbiology 11


Diagnosis of biofilm infections N.B.S. Silva et al.

resistance. Int J Gen Med 11, 25–32. https://doi.org/10. study of antimicrobial activity on Klebsiella Pneumoniae
2147/IJGM.S153268. biofilms in endotracheal tubes. J Chemother 31, 202–208.
Neu, T.R. and Lawrence, J.R. (2015) Innovative techniques, https://doi.org/10.1080/1120009X.2019.1601801.
sensors, and approaches for imaging biofilms at different Schlafer, S. and Meyer, R.L. (2017) Confocal microscopy
scales. Trends Microbiol 23, 233–242. https://doi.org/10. imaging of the biofilm matrix. J Microbiol Methods 138,
1016/j.tim.2014.12.010. 50–59. https://doi.org/10.1016/j.mimet.2016.03.002.
Nielsen, S.M., Penstoft, L.N. and Nørskov-Lauritsen, N. (2019) Shahmoradi, M., Faridifar, P., Shapouri, R., Mousavi, S.F.,
Motility, biofilm formation and antimicrobial efflux of Ezzedin, M. and Mirzaei, B. (2019) Determining the
sessile and planktonic cells of Achromobacter xylosoxidans. biofilm forming gene profile of Staphylococcus aureus
Pathogens 8, 14. 10.3390. clinical isolates via multiplex colony PCR method. Rep
Ommen, P., Zobek, N. and Meyer, R.L. (2017) Quantification Biochem Mol Biol 7, 181.
of biofilm biomass by staining: non-toxic safranin can Shukla, S.K. and Rao, T.S. (2017) An improved crystal violet
replace the popular crystal violet. J Microbiol Methods 141, assay for biofilm quantification in 96-well microtitre plate.
87–89. https://doi.org/10.1016/j.mimet.2017.08.003. Biorxiv 100214, https://doi.org/10.1101/100214.
Ordonez, A.A., Sellmyer, M.A., Gowrishankar, G., Ruiz- Singh, S., Singh, S.K., Chowdhury, I. and Singh, R. (2017)
Bedoya, C.A., Tucker, E.W., Palestro, C.J., Hammoud, Understanding the mechanism of bacterial biofilms
D.A. and Jain, S.K. (2019) Molecular imaging of bacterial resistance to antimicrobial agents. Open Microbiol J 11,
infections: overcoming the barriers to clinical translation. 53–62. https://doi.org/10.2174/1874285801711010053.
Sci Transl Med 11, 508. https://doi.org/10.1126/scitra Singhal, N., Kumar, M., Kanaujia, P.K. and Virdi, J.S. (2015)
nslmed.aax8251. MALDI-TOF mass spectrometry: an emerging technology
Osman, K., Orabi, A., Elbehiry, A., Hanafy, M.H. and Ali, for microbial identification and diagnosis. Front Microbiol
A.M. (2019) Pseudomonas species isolated from camel 6, https://doi.org/10.3389/fmicb.2015.00791.
meat: quorum sensing-dependent virulence, biofilm Solati, S.M., Tajbakhsh, E., Khamesipour, F. and Gugnani,
formation and antibiotic resistance. Future Microbiol 14, H.C. (2015) Prevalence of virulence genes of biofilm
609–622. https://doi.org/10.2217/fmb-2018-0293. producing strains of Staphylococcus epidermidis isolated
Otto, A., Bernhardt, J., Hecker, M. and Becher, D. (2012) from clinical samples in Iran. AMB Express 5, 47. https://
Global relative and absolute quantitation in microbial doi.org/10.1186/s13568-015-0134-3.
proteomics. Curr Opin Microbiol 15, 364–372. https://doi. Stepanovic, S., Vukovic, D., Hola, V., Bonaventura, G.D.,
org/10.1016/j.mib.2012.02.005. 
Djukic, S., Cirkovic, I. and Ruzicka, F. (2007)
Park, H.E.H., Jung, Y. and Lee, S.J.V. (2017) Survival assays Quantification of biofilm in microtiter plates: overview of
using Caenorhabditis elegans. Moll Cells 40, 90. https://doi. testing conditions and practical recommendations for
org/10.14348/molcells.2017.0017. assessment of biofilm production by staphylococci. Apmis
Peeters, E., Nelis, H.J. and Coenye, T. (2008) Comparison of 115, 891–899. https://doi.org/10.1111/j.1600-0463.2007.a
multiple methods for quantification of microbial biofilms pm_630.x.
grown in microtiter plates. J Microbiol Methods 72, Stoica, P., Chifiriuc, M.C., Rapa, M. and Lazar, V. (2017).
157–165. https://doi.org/10.1016/j.mimet.2007.11.010. Overview of biofilm-related problems in medical devices.
Percival, S.L. (2017) Importance of biofilm formation in In Deng, Y. and Lv, W., eds. Biofilms and Implantable
surgical infection. Br J Surg 104, e85–e94. https://doi.org/ Medical Devices, pp. 3–23. Oxford, UK: Woodhead
10.1002/bjs.10433. Publishing.
Pereira, F.D., Bonatto, C.C., Lopes, C.A., Pereira, A.L. and Sugimoto, S., Okuda, K.I., Miyakawa, R., Sato, M., Arita-
Silva, L.P. (2015) Use of MALDI-TOF mass spectrometry Morioka, K.I., Chiba, A., Yamanaka, K., Ogura, T. et al.
to analyse the molecular profile of Pseudomonas aeruginosa (2016) Imaging of bacterial multicellular behaviour in
biofilms grown on glass and plastic surfaces. Microb biofilms in liquid by atmospheric scanning electron
Pathog 86, 32–37. https://doi.org/10.1016/j.micpath.2015. microscopy. Sci Rep 6, 1–13. https://doi.org/10.1038/sre
07.005. p25889.
Qu, Y., Thissen, H., McGiffin, D.C. and Peleg, A.Y. (2017) Vuotto, C., Longo, F., Pascolini, C., Donelli, G., Balice, M.P.,
Optimizing microplate biofilm assays to screen anti- Libori, M.F., Tiracchia, V., Salvia, A. et al. (2017) Biofilm
infective surfaces. Trends Biotechnol 35, 3–5. https://doi. formation and antibiotic resistance in Klebsiella
org/10.1016/j.tibtech.2016.09.001. pneumoniae urinary strains. J Appl Microbiol 123,
Rajapaksha, P., Elbourne, A., Gangadoo, S., Brown, R., 1003–1018. https://doi.org/10.1111/jam.13533.
Cozzolino, D. and Chapman, J. (2019) A review of Wang, S., Yu, H. and Wickliffe, J.K. (2011) Limitation of the
methods for the detection of pathogenic microorganisms. MTT and XTT assays for measuring cell viability due to
Analyst 144, 396–411. superoxide formation induced by nano-scale TiO2. Toxicol
Ruiz, J., Sanjuan, E., Amaro, C., Gordon, M., Villarreal, E., in Vitro 25, 2147–2151. https://doi.org/10.1016/j.tiv.2011.
Castellanos-Ortega, A. and Ramirez, P. (2019) In vitro 07.007.

12 Journal of Applied Microbiology © 2021 The Society for Applied Microbiology


N.B.S. Silva et al. Diagnosis of biofilm infections

Washio, J. and Takahashi, N. (2016) Metabolomic studies of Developments in Microbial Biotechnology and
oral biofilm, oral cancer, and beyond. Int J Mol Sci 17, Bioengineering: Microbial Biofilms, pp. 15–28. https://doi.
870. org/10.1016/B978-0-444-64279-0.00002-5.
Xu, Z., Liang, Y., Lin, S., Chen, D., Li, B., Li, L. and Deng, Y. Yamaguchi, M. and Yoshida, H. (2018) Drosophila as a model
(2016) Crystal violet and XTT assays on Staphylococcus organism. In Yamaguchi, M., ed. Drosophila Models for
aureus biofilm quantification. Curr Microbiol 73, 474–482. Human Diseases, pp 1–10. Singapore: Springer.
https://doi.org/10.1007/s00284-016-1081-1. Zhang, B. and Powers, R. (2012) Analysis of bacterial biofilms
Yadav, M.K., Vidal, J.E. and Song, J.J. (2020) Microbial using NMR-based metabolomics. Future Med Med 4,
biofilms on medical indwelling devices. In New and Future 1273–1306. https://doi.org/10.4155/fmc.12.59.

Journal of Applied Microbiology © 2021 The Society for Applied Microbiology 13

You might also like