Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Manuscript File - Clean Click here to view linked References

RESEARCH ARTICLE

A Mild catalyzed imino Diels-Alder reaction for the synthesis of N-(2-


(o-tolyl)-1,2,3,4-tetrahydroquinoline-4-yl)formamide derivatives as
regulators of Quorum Sensing in Pseudomona Aeruginosa

Leidy J. García Maza1, Dayanna F. Orosco Flórez1, Arturo René Mendoza Salgado1, Wendy Rosales2, Evelyn Mendoza-Torres3
and Carlos Mario Meléndez*1
1
Faculty of Basic Science, Molecular Biology Research Laboratory, Organic and BiomedicalChemistry Research Group, Universidad del Atlántico,
Barranquilla, Colombia.
2
Faculty of Exact and Natural Sciences, Biomedicine Advanced Research Group, Universidad Libre, Barranquilla, Colombia.
3
Faculty of Health Sciences, Biomedicine Advanced Research Group, Universidad Libre, Barranquilla, Colombia.

Bacterial resistance is one of the major global public health problems. In addition, virulence factors promoted multiple drug resistance
through cell-to-cell communications by molecules within microbial communities, denominated Quorum Sensing (QS). The regulation of
QS by small molecules appears as a therapeutic alternative through the use of synthetic analogs of agonists. The present work developed the
construction of a series of derivatives of N-(2-(o-tolyl)-1,2,3,4-tetrahydroquinoline-4-yl) formamide (1a-h) via Povarov-type sequential
reactions to deliver the tetrahydroquinoline (THQ) derivatives in moderate to good yields (24-78%). To evaluate the potential in molecular
diversity of THQ-type autoinducers, a study of these agents as potential inhibitors of bacterial growth and virulence factors involved in QS
systems on P. aeruginosa was carried out both as antibacterial evaluation on E. coli strain. THQs showed significant activity values for
molecules with halogen substituents at the C-6 position (-F, -Cl, and I), ), revealing at a concentration of 75 µg/mL a percentage inhibition
between 27-40%. Furthermore, autoinducers with halogen substitutes in phenotypic studies on P. aeruginosa showed good results. Also,
they exhibited weak formation of both biofilm and low pyocyanin formation, which are indicators of virulence in P. aeruginosa. Overall,
the results suggest that compound N-(6-fluor-2-(o-tolyl)-1,2,3,4-tetrahydroquinoline-4-yl)formamide (1a) stands out as the most
representative within this series of THQ derivatives.

Keywords: Antibacterial resistance, Quorum Sensing, Synthetic inhibitors, Tetrahydroquinolines.

1. Introduction promising approach for the treatment of bacterial infections.


[8]. Significant efforts have focused on the development of
Resistance of bacterial pathogens to antimicrobials is a
new strategies for the treatment of biofilm infections,
global problem [1],[2]. Conventional therapies and numerous
particularly targeting the processes of cell dispersion in
currently available antibiotics have proven ineffective
biofilm [9]. Other approaches include the use of
against clinically relevant strains of both Gram-negative and
antimicrobial peptides [10], bacteriophage systems [9], and
Gram-positive bacteria, leading to the emergence of several
inhibitors of Quorum Sensing [14], among others.
mechanisms of multi-drug resistance [3], [4]. Gram-negative
pathogens such as Pseudomonas aeruginosa and Escherichia
coli concerning due to their recurrent resistance processes,
which are often more severe compared to Gram-positive
pathogens [5]–[10]. The main challenge in combating
antibiotic resistance lies in the ineffectiveness of current
antibiotic therapy against biofilms established by bacteria
[11]–[15]. Virulence factors such as the formation of
biofilms protect the microbial communities ensuring the
communication and development of the bacterial population,
these factors are promoted through a cell-to-cell
communication system called Quorum Sensing (QS) [16]– Fig. 1. Native autoinducers in Quorum sensing in P. aeruginosa: 3-
[18], a mechanism responsible for the regulation of virulence o-C12-HSL, C4-HSL, PQS, and HHQ. The 2-alkyl-4-quinolones
genes depending on the bacterial density [15], [19], [20]. As molecules share an analogous nucleus with the
tetrahydroquinolines.
a result, targeting biofilm growth has emerged as a

https://doi.org/10.26434/chemrxiv-2023-7f4vv ORCID: https://orcid.org/0000-0003-0637-5885 Content not peer-reviewed by ChemRxiv. License: CC BY-NC-ND 4.0
2

The reaction between anilines (2), o-tolualdehyde (3), and N-


Therefore, comprehending, controlling, and/or regulating vinyl formamide (4) in the presence of p-TsOH 10 mol% p-
these bacterial chemical communication systems hold the toluenesulfonic acid, and solvents such as ethanol and
potential to establish new objectives for developing acetonitrile at room temperature provides ready access to (2-
alternative or complementary treatments to conventional (o-tolyl)-1,2,3,4-tetrahydroquinoline-4-yl)formamide
antibiotic agents [8], [18], [21], the development of these derivatives (1a-h) (Scheme 1). While striving to extend the
strategies prevents or limits the formation of biofilms scope of this transformation, we have discovered that the use
through the use of synthetic analogs of natural autoinducers at high temperatures or carrying out the reaction in one pot
[9], [16], [22]–[24]. (Tetrahydro)quinoline (THQ) would cause the reaction to deviate to a different course,
derivatives represent a significant strategy in the molecular forming several degradation products.
design of synthetic analogs of natural autoinducers [25]–
[29]. The structural diversity of these heterocyclic Table 1. Catalyst and solvent screening for imino Diels-Alder
compounds [30], [31] along with their shared nucleus reaction of substituted anilines (2), o-tolualdehyde (3), and N-vinyl
analogous to the native autoinducers PQS corresponding to formamide (4).
the HHQ signals [23], [24], [32]–[34] (Fig. 1), make them as
Entry Catalyst Solvent (b/c) Temp Yield
a promising antimicrobial agent against resistant pathogens,
(°C) (%)
in addition to the exploration of new systems that can
modulate QS circuits [14], [21], [35]–[37]. On the other 1 ZnCl2d EtOHb,c r.t 30
hand, although THQ derivatives have been widely reported 2 BiCl3e,f CH2Cl2/MeCN r.t 42
for their broad spectrum of antibacterial activities [38]–[41], 3 ZnCl2e EtOH/MeCN 80-120g nr
their potential as QS inhibitors has not been fully developed 4 BiCl3d,f CH2Cl2/MeCN r.t 42
yet [32]. Based on the above, searching for new Micellar
5 MeOHi r.t 37
antimicrobials and using effective synthetic routes to obtain catalysis
THQ derivatives with potent biological activities is 6 BiCl3d EtOH/MeCN r.t 50
imperative. The imino-Diels-Alder reaction (iDA) emerge as 7 Ce(SO4)2d EtOH/MeCN r.t nr
an effective methodology [31], [42]–[44] for HOAc 1
tetrahydroquinoline compounds. [30], [42] The iDA reaction 8 EtOH/MeCN r.t 18
mmol
offer several advantages as readily available reagents [45], 9 TsOHe EtOH/MeCN r.t 71
high regioselectivity and stereospecificity [46], and high Amberlyst
yields, among others. EtOH/PEG
10 15 (40% r.t 27
600
w/w)
a
This work aims to synthesize new non-native autoinducers Catalyst in second step; b Solvent in first step; c Solvent in second step; d 5
% mol; e 10 % mol; f Na2SO4 (2.0 equiv, 4.0 mmol); g refluxing EtOH; h 15
using the iDA reaction, via a simple and versatile protocol, mL SDS-H2O/HCl 0,1 M; i one-pot reaction; r.t, room temperature; nr, no
with readily available arylamines, o-tolualdehyde, and N- reaction.
vinyl formamide. Additionally, the effectiveness of the
synthesized compounds will be evaluated in the regulated Overall, moderate yields (24-78%) were obtained during
QS by observing phenotypic changes in E. coli and P. Diels-Alder imino cycloaddition, the compounds with
aeruginosa strains. This research offers a new perspective on electron-donating groups such as -CH3 (71%) and -OMe
the significance of exploring the molecular diversity of (78%) showed the best global yields. Moreover, compounds
synthetic analogs of autoinducers such as with electron-withdrawing groups e.g., -F (56%), -Cl (44%),
tetrahydroquinoline compounds, as a starting point for and -I (56%) exhibited moderate yields and do not reveal a
further investigations on QS systems and the potential marked trend as atomic radius increases and
application as therapeutic agents against drug-resistant
electronegativity decreases. The results obtained from the
bacterial strains.
reaction yields are consistent with those reported in studies
2. Chemistry: synthesis of the 2-(o-tolyl)-1,2,3,4- related to the synthesis of tetrahydroquinolines by the imino
tetrahydroquinoline Diels-Alder reaction with the use of arylamines and aromatic
aldehydes, with moderate to good yields [51].
The iDA sequential methodology has been extensively
employed to explore and optimize reaction conditions using
The yield of the product in the reaction can be attributed to
readily available and commercially accessible substrates
the easy formation and stability of the imine intermediate
(Table 1). Diverse catalysts [31], [47], [48] and solvents
(5). Additionally, aromatic amines with electron-donating
have been analyzed to identify the most efficient reaction
groups have demonstrated similar reactivity, contributing to
conditions, various reaction media as micellar catalysis with
the yield of the reaction. These groups donate some of their
SDS-H2O/HCl 0. 1 M used in green chemistry, as well as
electron density to the aromatic ring of aniline, which allows
solvents such as PEG-400 and glycerol [49], and catalysts
most of the electron density to be concentrated in the
such as Amberlyst 15 [50] have also been used. The optimal
nitrogen atom leading to imine formation more easily than
reaction conditions for obtaining the desired product were
with a group that reduces electron density in the molecule
identified to be achieved using the catalyst p-TsOH and
around the nitrogen atom. Moreover, arylamines with
EtOH/MeCN as solvent (entry 9).
electron-donating bulky groups (1b, 1h) do not react
completely with the aromatic aldehyde to give the desired

https://doi.org/10.26434/chemrxiv-2023-7f4vv ORCID: https://orcid.org/0000-0003-0637-5885 Content not peer-reviewed by ChemRxiv. License: CC BY-NC-ND 4.0
3

intermediate possibly due to the steric hindrance. This causes of N-(2-(o-tolyl)-1,2,3,4-tetrahydroquinoline-4-yl)formamide


the electron density to be partially concentrated on the 1a-h were structurally characterized using spectroscopy
nitrogen atom and the complete formation of the imine does techniques such as IR, NMR one-dimensional (1H, 13C) and
not occur, although the reaction time is prolonged. The series two-dimensional (COSY, HMBC, HSQC, DEPT 135).

Scheme 1. Synthesis of N-(2-(o-tolyl)-1,2,3,4-tetrahydroquinoline-4-yl)formamide (1a-h) under optimal conditions.

3. Biology
Bacterial growth curves show the maximum absorbance
3.1 Antibacterial activity recorded at 8 hours. Therefore, the mean absorbances of the
culture medium obtained from 3 independent experiments
The inhibitory capacity of tetrahydroquinoline (1a-h) was were calculated and as a result the inhibition percentages of
assessed against the Gram-negative strains of clinical the THQ compounds (1a-h) for each bacterial strain were
importance E. coli and P. aeruginosa [13]. Growth control acquired. The substituted 6-fluor compound 1a, followed by
experiments were conducted to observe the initial phases of the halogenated compounds 1d and 1f showed the greatest
growth for each concentration (10, 20, 35, 50, 75 µg/mL) of decrease in absorbance at a concentration of 75 µg/mL for
THQ derivatives Figure 2. The latency phase corresponds to both P.aeruginosa and E. coli, in an inhibition range of 20-
the adaptation period (first two hours), followed by the 40 %. The absorbance values obtained determine the
exponential phase characterized by cell division (last six inhibition percentages of compounds evaluated (Figure 3)
hours) [52]. The results demonstrated a direct relationship which evidence higher values for the compounds mentioned
between the P. aeruginosa and E. coli growth and the above. Whereas compound 1e does not induce a decrease in
concentration of THQ derivatives, showing consistent trends the growth of both microorganisms.
in growth control for both bacterial strains. All compounds
exhibited growth inhibition effects as early as 4 hours, Figure 3 illustrates a progressive increase in the inhibition
particularly at concentrations of 50 and 75 µg/mL. percentage as the concentration of the compounds increases.
Compared those results with the positive control However, it is important to identify the minimum
(Gentamycin 10 µg/mL), we notice the remarkable concentration of tetrahydroquinolines compounds at which
effectiveness and potency of this drug [40], [53], [54]. growth inhibition initiates. To determine it, the average
Regarding cytotoxicity control (yellow line), it is absorbance of the cultures at 8 hours of incubation was
corroborated that the solvent used (DMSO) for the calculated for each concentration in µg/mL. Subsequently, a
preparation of stock solutions of the derivatives did not post hoc Tukey test was performed to identify significant
influence the bacteria growth. statistical differences between the group of concentrations
and growth control. This analysis allows the determination

https://doi.org/10.26434/chemrxiv-2023-7f4vv ORCID: https://orcid.org/0000-0003-0637-5885 Content not peer-reviewed by ChemRxiv. License: CC BY-NC-ND 4.0
4

of minimum inhibitory concentration (MIC), finding that the tetrahydroquinoline derivatives with halogenated
MIC inhibits bacterial growth for most compounds starting substituents at the C-6 position [55][39] exhibited higher
at 10 and 20 µg/mL with a range of p < 0,05-0,0001. inhibition percentages compared to compounds with
electron-donating groups such as
The obtained results are consistent with the trend observed in
previous structure-activity relationship studies, where

a b

P. aeruginosa E. coli P. aeruginosa E. coli


1.2 1.2 1.2 1.2
Abs690 (AU)

Abs660 (AU)

Abs690 (AU)

Abs660 (AU)
0.8 0.8 0.8 0.8

0.4 0.4 0.4 0.4

0.0 0.0 0.0 0.0


0 2 4 6 8 0 2 4 6 8 0 2 4 6 8 0 2 4 6 8
Time (hours) Time (hours) Time (hours) Time (hours)

c d
P. aeruginosa E. coli P. aeruginosa E. coli
1.2 1.2 1.2 1.2
Abs690 (AU)

Abs660 (AU)

Abs690 (AU)

Abs660 (AU)
0.8 0.8 0.8 0.8

0.4 0.4 0.4 0.4

0.0 0.0 0.0 0.0


0 2 4 6 8 0 2 4 6 8 0 2 4 6 8 0 2 4 6 8
Time (hours) Time (hours) Time (hours) Time (hours)

e f
P. aeruginosa E. coli P. aeruginosa E. coli
1.2 1.2 1.2 1.2
Abs690 (UA)

Abs660 (AU)

Abs690 (AU)

0.8 0.8 0.8 Abs660 (AU) 0.8

0.4 0.4 0.4 0.4

0.0 0.0 0.0 0.0


0 2 4 6 8 0 2 4 6 8 0 2 4 6 8 0 2 4 6 8
Time (hours) Time (hours) Time (hours) Time (hours)

g h
P. aeruginosa E. coli P. aeruginosa E. coli
1.2 1.2 1.2 1.2
Abs690 (UA)

Abs660 (UA)

Abs690 (UA)

Abs660 (UA)

0.8 0.8 0.8 0.8

0.4 0.4 0.4 0.4

0.0 0.0 0.0 0.0


0 2 4 6 8 0 2 4 6 8 0 2 4 6 8 0 2 4 6 8
Time (hours) Time (hours) Time (hours) Time (hours)

10 µg/mL 20 µg/mL 35 µg/mL 50 µg/mL 75 µg/mL C+ gentamicin 10 µg/mL

Cytotoxicity (DMSO) Growth control

Figure 2. Growth curves of P. aeruginosa and E. coli when exposed to different concentrations of N-(2-(o-tolyl)-(1,2,3,4-
tetrahydroquinoline-4-yl)formamide derivatives using the absorbance variable as a growth indicator. (n=3, independent experiments). a)
compound 1a, b) compound 1b, c) compound 1c, d) compound 1d, e) compound 1e, f) compound 1f, g) compound 1g y h) compound 1h.

-Me and -OMe (F > I > Cl > Me > H > C4H10 (isopropyl)). breakage of bacterial chromosomes [56]. Therefore, it is
This is possibly due to atoms such as fluorine enhancing the noteworthy that the compound N-(6-fluor-2-(o-tolyl)-1,2,3,4-
cellular barrier penetration of the compounds and might tetrahydroquinoline-4-yl)formamide 1a is the most
effectively inhibit the DNA gyrase complex, resulting in the representative of the series of THQ derivatives. It exhibited

https://doi.org/10.26434/chemrxiv-2023-7f4vv ORCID: https://orcid.org/0000-0003-0637-5885 Content not peer-reviewed by ChemRxiv. License: CC BY-NC-ND 4.0
5

the highest inhibition percentages on the studied strains E. 10 µg/mL with p < 0,0001). Other compounds show
coli (35,9%) and P. aeruginosa (39,1%), being the MIC = 10 moderate antibacterial activity such as 1c and 1g on P.
µg/mL where the inhibition of bacterial growth on both aeruginosa likewise compound 1f when was evaluated on
pathogens begins with con p < 0,0001. In addition, the both pathogens. Overall, the analyzed compounds exhibited
compound N-(6-chloro-2-(o-tolyl)-1,2,3,4- lower antibacterial activity compared to the reference drug
tetrahydroquinoline-4-yl) formamide 1d showed a similar (positive control), as indicated by the yellow bars in Figure
effect but in this case only on P. aeruginosa (36,8%, MIC = 3.

68,7
80

70,1

70,3

68,3

67,4

68,3

68,5
65,3
60
% Inhibition

39,1

36,8
35,4

35,6
34,7

34,5
32,8
40

26,2
26,2

25,9
22,1
22,1

21,7
20,9

19,1
19,0

18,2

17,6
16,7

17,6
17,4
15,8

15,0

16,7
14,6

14,8

14,5
13,6

13,3

12,8
11,9
12,5

11,2
20

9,0

9,4
8,7

7,5
6,8
4,9
3,3

0
1a 1b 1c 1d 1e 1f 1g 1h
Compound

80,7
80,7

b
78,8

71,7

71,5

71,8

68,49
68,49
80

60
% Inhibition

35,9

32,8

40
26,9
26,4

21,56

20,32
20,76
18,12
20,9

21,5

20,2

20,7
19,3

16,78

18,5
13,9
16,5

15,7

14,2

13,03
17,2

10,85
12,7

12,8

11,2
12,1

20
11,2
8,50
9,50

8,80

8,10

6,86

7,34
6,12

7,02
6,34

5,53
5,60

5,72
4,25

0
1a 1b 1c 1d 1e 1f 1g 1h
Compound
10 µg/mL 20 µg/mL 35 µg/mL 50 µg/mL 75 µg/mL
C+ gentamicin 10 µg/mL

Figure 3. Growth inhibition of a) P. aeruginosa and a) E. coli, after exposure to different concentrations of THQ derivatives (1a-f).

Indeed, certain studies propose that antibacterial agents can


interfere with the bacterial transcription process, preventing 3.2 Biofilm Formation Index (BFI)
RNA or DNA transcription [57]. Moreover, DNA gyrase is P. aeruginosa is associated with biofilm-forming bacteria,
the primary target for most Gram-negative bacteria, which often cause resistance to antibiotics used to treat
including the opportunistic pathogens P. aeruginosa and E. acquired clinical infections. The mechanisms of biofilm
coli [56]. Therefore, the antibacterial activity exhibited by formation in P. aeruginosa are regulated by Quorum
THQ compounds with electron-withdrawing groups is Sensing, mainly by 3 systems las, rhl, and pqs, the latter
attributed to the interaction with the DNA polymerase. These being mediated by signaling molecules 2-heptyl-3-hydroxy-
molecules facilitate cellular penetration and contribute to the 4(1H)-quinolone (PQS) [59][60]. However, the mechanism
inhibition of the DNA gyrase complex [56]. The ability of of action of PQS in promoting biofilm formation remains
bacteria agents to pass through the bacterial envelope and unclear [59], [61], [62]. There is potential for PQS analogs to
reach their biological activity is crucial for their efficacy. serve as biofilm inhibitors. Despite recent studies on the
The moderate activity exhibited by the synthesized effect of tetrahydroquinolines compounds on P. aeruginosa
compounds may be due to their ability to penetrate the cell biofilm formation are lacking, it is possible that analogous of
barrier and, by the efficiency of efflux pumps that decrease the native autoinducer PQS as N-(2-(o-tolyl-(1,2,3,4-
intracellular concentration of bacterial agents in the tetrahydroquinoline-4-yl)formamide (1a-h), could act as
cytoplasm [58]. inhibitors of biofilm formation. First, absorbance values

https://doi.org/10.26434/chemrxiv-2023-7f4vv ORCID: https://orcid.org/0000-0003-0637-5885 Content not peer-reviewed by ChemRxiv. License: CC BY-NC-ND 4.0
6

were obtained at the different concentrations evaluated after 35, and 75 μg/mL, i.e., it has the greatest decrease in biofilm
24 hours of incubation with the bacterial strain for formation in P. aeruginosa. This result is consistent with the
subsequent analysis of the biofilm inhibition capacity of observed inhibition of bacterial growth. Compounds with
possible THQ autoinducers (Figure 4). more than one or bulky substituents in the aromatic ring (1b
and 1h) increase biofilm formation; including compound 1e
The results showed a strong tendency for the reduction of without electron-withdrawing or -donating groups in its
biofilm production (decrease in absorbance) with the structure.
progressive increase of the concentration of synthetic
autoinducers (Figure 4) [33]. It observes the negative The effect observed by the synthetic analogs (THQ) at the
control (bacterial strain without autoinducers) with a lowest concentration (10 μg/mL) is particularly intriguing. It
progressive increase in absorbance measurements and then is noteworthy that some of the studied molecules exhibit a
remaining constant but with a significant difference with the biofilm formation rate very similar to the negative control,
assays that contained the autoinducers analogous to the indicating no significant influence on the inhibition of
different concentrations. biofilm formation. However, as the concentration of the
compounds increases, a decrease in bacterial biofilm
formation becomes evident, suggesting a potential alteration
0.8
1a in bacterial communication circuits mainly targeting
0.7 1b compound 1a. Recent research has found that the amount of
substituents in the phenyl ring influences the inhibitory
Abs570 (AU)

1c
0.6 activity of biofilm formation [33]. This explains the low
1d
0.5 1e activity (high rates of biofilm formation) of the compound
1f 1h because the aromatic ring is replaced more than once. In
0.4 1g addition, increases in the length of the substituted group
1h chains in the benzene ring drastically increase the biofilm
0.3
0 10 20 30 40 50 60 70 Control (-) formation, as in the case of the synthesized antagonist 1b.
Concentration (g/mL)
It is observed in Table 2 that more than 98% of the
concentrations of the autoinducers present BFI ≥ 1.0 being
Figure 4. Recorded absorbance after incubation for 24 hours of o- classified as moderate to strong biofilm formers and, BFI <
tolyl (1,2,3,4-tetrahydroquinoline-4-yl)formamide derivatives (1a-f) 1.0 is considered as weak formers, according to studies
on P. aeruginosa strain. Control (-), growth control of the bacterial performed on biofilm formation [63]. Therefore, it is
strain without autoinducer.
evidenced in this study that the evaluated molecules induce
The optical density results expressed in terms of the Biofilm the P. aeruginosa bacterial strain to develop moderate to
Formation Index (BFI) allow us to classify the ability of the weak biofilm indices at 75 µg/mL with p < 0.0001, i.e., these
compounds (1a-h) to modify the capacity of biofilm analogs can decrease biofilm formation by up to 50% if we
formation on P. aeruginosa (Table 2). Moreover, compound compare with the BFI data of C- (100% biofilm formed by
1a with a halogen substituent at the C-6 (-F) position has the the bacterial strain). These results open the possibility of
lowest biofilm formation index compared to the other future molecular studies to determine the specific genes that
synthetic autoinducers evaluated at concentrations of 10, 20, are expressed for this type of virulence mechanism.

Table 2. Biofilm formation Index (BFI) of P.aeruginosa of each of the autoinducers of N-(2-(o-tolyl)-1,2,3,4-tetrahydroquinoline-4-yl)
formamide at the concentration evaluated. (N = 3, number of independent essays).

Synthetic Concentration (µg/mL)


autoinducer
10 20 35 50 75 (C-)
1a 1,50 1,24 1,19 1,08 0,81 1,95
1b 1,93 1,54 1,45 1,19 1,09 1,95
1c 1,64 1,50 1,20 1,04 0,97 1,95
1d 1,62 1,40 1,33 1,15 0,89 1,95
1e 1,94 1,52 1,32 1,24 1,09 1,95
1f 1,64 1,24 1,24 1,20 0,96 1,95
1g 1,63 1,39 1,43 1,04 0,95 2,01
1h 1,77 1,55 1,36 1,08 1,05 1,83

3.3 Pyocyanin Production

https://doi.org/10.26434/chemrxiv-2023-7f4vv ORCID: https://orcid.org/0000-0003-0637-5885 Content not peer-reviewed by ChemRxiv. License: CC BY-NC-ND 4.0
7

Pyocyanin is an important virulence factor of P. aeruginosa the concentration of the evaluated autoinducers increases.
[33]. This is one of the phenazine compounds with redox Table 3 shows the average absorbances for each of the
activity produced and secreted by this pathogen. Although concentrations of the molecules tested, showing a decrease in
this exotoxin has a wide range of toxic effects on different pyocyanin formation of 44% and 43% at 75 µg/mL for
host cells, its different toxicity originates from the compounds 1a and 1d, respectively. Hence, the effectiveness
production of reactive oxygen species (ROS) capable of of halogenated substituents is observed in regulating some
causing significant oxidative stress [64], [65]. Moreover, the virulence factors such as pyocyanin production and biofilm
exoproduct pyocyanin (PCN) is a zwitterion that easily formation. Statistical analysis by Tuckey's test indicated a
penetrates biological membranes [66]. Therefore, pyocyanin significant difference between the means of the data versus
is an essential component for complete virulence in a variety growth control, starting at 10 µg/mL with p < 0.05 for 1a
of Pseudomonas aeruginosa infection models [67], [68]. and p < 0.01 for 1g. Furthermore, a significant statistical
difference with p < 0.0001 was observed at 35 µg/mL for all
The formation of PCN in the culture of P. aeruginosa was synthetic autoinducers studied in this assay.
determined by evaluating the molecules that presented the
lowest rates of biofilm formation which correspond to 1a, As the compounds (1a-h) show an effect on bacterial growth
1c, 1d, 1f, and 1g. The absorbance of the cultures at 600 nm inhibition < 40%, it is suggested that the synthetic
was recorded after 20 hours of incubation for calculations of autoinducers do not cause the death of the evaluated P.
pyocyanin production. As phenotypic identification, the aeruginosa pathogen. However, they regulate some
production of pyocyanin is responsible for the blue-green virulence factors that generate resistance to standard
color characteristic of P. aeruginosa, observing a decrease as antibiotics.

Table 3. Mean absorbances of the culture medium (an indicator of pyocyanin formation) after exposure of the P. aeruginosa inoculum to
each concentration at 20 hours of incubation (n=3). The means ± Standard Deviation (SD) of each of the recorded absorbances are shown.
Growth control as bacterial strain without autoinducer.

Compound 1a 1c 1d 1f 1g

Control 1,16(±0,03) 1,10 (±0,03) 1,11 (±0,03) 1,14 (±0,03) 1,18(±0,03)


10 µg/mL 1,04(±0,004) 1,06(±0,014) 1,05(±0,02) 1,074(±0,03) 1,04(±0,04)
20 µg/mL 1,02(±0,024) 1,02(±0,021) 1,02(±0,02) 1,03(±0,013) 1,01(±0,03)
35 µg/mL 0,946(±0,04) 0,826(±0,043) 0,991(±0,01) 0,951(±0,04) 0,943(±0,002)
50 µg/mL 0,843(±0,039) 0,801(±0,033) 0,93(±0,04) 0,876(±0,04) 0,812(±0,04)
75 µg/mL 0,732(±0,031) 0,797(±0,011) 0,749(±0,04) 0,801(±0,03) 0,791(±0,01)
The reagents and other materials were acquired from
certified chemical companies. Dimethyl sulfoxide (DMSO)
4 Materials and Methods was purchased from Merck. Müeller Hinton broth (MH
4.3 General Information broth) and Müeller Hinton agar (MH agar) were obtained
from Merck KGaA (Darmstadt, Germany). Gentamicin
All reagents and solvents used were reactive grades reactive grade Sigma Aldrich. The bacterial strains were
purchased from (Sigma Aldrich and Merck). Column from the American Type Culture Collection (ATCC):
chromatography was performed using spherical silica gel 70 Pseudomonas aeruginosa (ATCC 27853) by Microbiologics
Å, 40–75 μm. One-dimensional (1H, 13C) and two- and Escherichia coli (ATCC 25922) by Sigma Aldrich.
dimensional (COSY, HMBC, HSQC, and DEPT 135) ALTA ELISA Plate Analyzer (405 – 630 nm) was used for
Nuclear Magnetic Resonance (NMR) were recorded on a the reading of absorbances (Universidad Libre, Barranquilla,
Bruker Avance-400 (400 MHz) spectrometer (Universidad Colombia).
Industrial de Santander, Bucaramanga, Colombia) with the
solvent resonance as the internal standard (CDCl3: δ = 7.26;). 4.4 Synthesis of N-(2-(o-tolyl)-1,2,3,4-
Infrared spectra (FT-IR) were measured on a tetrahydroquinoline-4-yl)formamide (1a-h)
Spectrophotometer FTIR-4700 + ATR PRO ONE
(Universidad del Atlántico, Barranquilla, Colombia). In a 50 mL round bottom ball were added 1 mmol of 2a-f
Melting points were measured on a Differential Scanning aniline, 1 mmol of 3 o-tolu aldehyde, and 10 mL EtOH, then
Calorimeter (DSC) (823) (Universidad del Atlántico, it was covered with a septum and subjected to constant
Barranquilla, Colombia). The reactions were carried out for agitation at room temperature. Once the 5a-h N-phenyl-2-(o-
the synthesis of 1a-h. tolyl)methanimine (observed by TLC) was performed, then
the reaction crude was transferred to a 100 mL round bottom
To determine the biological activity of the derivatives of N- ball, and the solvent was evaporated at reduced pressure. To
(2-(o-tolyl)-1,2,3,4-tetrahydroquinoline-4-yl) formamide the 100 mL round ball which contained 1 mmol of N-phenyl-
(1a-h), three types of assays were performed to analyze the 2-(o-tolyl)methanimine (without purification) was added 10
regulation of these systems using the synthesized molecules. mol% of p-toluenesulfonic acid (p-TsOH), 1 mmol of 4 N-
vinyl formamide and 20 mL MeCN, then it was covered with
a septum and subjected to constant agitation at room

https://doi.org/10.26434/chemrxiv-2023-7f4vv ORCID: https://orcid.org/0000-0003-0637-5885 Content not peer-reviewed by ChemRxiv. License: CC BY-NC-ND 4.0
8

temperature. Product formation was confirmed by TLC. The


solvent of the reaction crude was evaporated at reduced 4.4.4 N-(6-chloro-2-(o-tolyl)-1,2,3,4-
pressure. The reaction mass was washed with brine and the tetrahydroquinoline-4-yl)formamide (1d)
organic phase was extracted with dichloromethane (3x20
mL) and deposited on anhydrous sodium sulfate (Na2SO4). White solid; yield: 0.22 g (0.73 mmol, 44%); mp: 150,82 °C.
The organic phase was concentrated under reduced pressure. IR (ATR): v max (cm-1) 3382, 3234, 3029, 2958, 2925, 2865,
The crude material was purified by column chromatography 1652, 1490, 1380, 1301, 1089, 754. NMR 1H (400 MHz,
(silica gel, hexane: ethyl acetate; in variable proportions) to CDCl3): δ 8.31 (s, 1H, (CO)H), 7.53 (d, J = 2.7 Hz, 1H, 6’-
provide the desired N-(2-(o-tolyl)-1,2,3,4- HAr), 7.26 – 7.20 (m, 3H, 3’,4’,5’-HAr), 7.15 (d, J = 1.6 Hz,
tetrahydroquinoline-4-yl)formamide 1a-h. 1H, 5-HAr), 7.04 (dd, J = 8.9, 2.0 Hz, 1H, 7-HAr), 6.52 (d, J =
8.5 Hz, 1H, 8-HAr), 5.81 (d, J = 9.3 Hz, 1H, N-H(CO)), 5.56
4.4.1 N-(6-fluor-2-(o-tolyl)-1,2,3,4- (ddd, J = 10.3, 10.0, 6.3 Hz, 1H, 4-Hax), 4.83 (dd, J = 10.9,
tetrahydroquinoline-4-yl)formamide (1a) 2.6 Hz, 1H, 2-Hax), 4.03 (s, 1H, N-H), 2.44 – 2.42 (m, 1H,
3b-Hec), 2.40 (s, 3H, 2’-CH3), 1.92 – 1.81 (m, 1H, 3a-Hax)
White crystals; yield: 0.28 g (0.98 mmol, 56%); mp: 148,76 ppm. NMR 13C (101 MHz, CDCl3): δ 161.1, 144.0, 140.3,
°C. IR (ATR): v max (cm-1) 3367, 3226, 3041, 2962, 2925, 135.2, 130.9, 128.5, 127.7, 127.0, 126.6, 125.5, 122.4, 122.1,
2869, 2755, 1650, 1502, 1454, 1382, 1261, 750. NMR 1H 115.8, 51.7, 44.9, 36.5, 19.1 ppm.
(400 MHz, CDCl3): δ 8.29 (s, 1H, (CO)H), 7.56 (d, J = 2.6
Hz, 1H, 6’-HAr), 7.26 – 7.20 (m, 3H, 3’,4’,5’-HAr), 6.93 – 4.4.5 N-(2-(o-tolyl)-1,2,3,4-tetrahydroquinoline-
6.89 (m, 1H, 5-HAr), 6.82 – 6.81 (m, 1H, 7-HAr), 6.52 (dd, J 4-yl)formamide (1e)
= 8.8, 4.7 Hz, 1H, 8-HAr), 5.92 (d, J = 9.3 Hz, 1H, N-
H(CO)), 5.57 (ddd, J = 10.0, 9.8, 6.1 Hz, 1H, 4-Hax), 4.79 Beige oil; yield: 0.21 g (0.79 mmol, 42%); mp: 110,95 °C.
(dd, J = 11.0, 2.5 Hz, 1H, 2-Hax), 3.92 (s, 1H, N-H), 2.42 IR (ATR): v max (cm-1) 3319, 3269, 3050, 3019, 2952, 2923,
(dd, J = 6.0, 2.4 Hz, 1H, 3b-Hec), 2.40 (s, 3H, 2’-CH3), 1.94 2863, 1663, 1603, 1476, 1381, 1310, 1247, 1038, 745. Peso
– 1.78 (m, 1H, 3a-Hax) ppm. NMR 13C (101 MHz, CDCl3): δ molecular: 266.34 g/mol. NMR 1H (400 MHz, CDCl3): δ
161.1, 141.8, 140.5, 135.2, 130.8, 127.6, 126.6, 125.5, 115.7, 8.28 (s, 1H, (CO)H), 7.56 (d, J = 6.3 Hz, 1H, 6’-HAr), 7.23 –
115.6, 115.4, 113.7, 113.5, 52.0, 45.1, 36.8, 19.1 ppm. 7.20 (m, 4H, 3’,4’,5’,5-HAr), 7.12 – 7.08 (m, 1H, 6-HAr), 6.77
– 6.73 (m, 1H, 7-HAr), 6.59 (dd, J = 8.0, 1.2 Hz, 1H, 8-HAr),
4.4.2 N-(8-isopropyl-2-(o-tolyl)-1,2,3,4- 5.88 (d, J = 9.3 Hz, 1H, N-H(CO)), 5.60 (td, J = 10.1, 5.9
tetrahydroquinoline-4-yl)formamide (1b) Hz, 1H, 4-Hax), 4.84 (dd, J = 10.9, 2.6 Hz, 1H, 2-Hax), 4.02
(s, 1H, N-H), 2.45 (dd, J = 6.0, 2.6 Hz, 1H, 3b-Hec), 2.41 (s,
White solid; yield: 0.12 g (0.39 mmol, 24%); mp: 162,98 3H, 2’-CH3), 2.07 (s, 1H, 3a-Hax) ppm. NMR 13C (101 MHz,
°C. IR (ATR): v max (cm-1) 3324, 3264, 3193, 3035, 2960, CDCl3): δ 161.2, 145.5, 140.8, 135.2, 130.8, 128.7, 127.6,
2927, 2869, 1671, 1596, 1544, 1494, 1247, 742. NMR 1H 127.3, 126.6, 125.6, 120.7, 118.0, 114.7, 51.7, 45.1, 37.0,
(400 MHz, CDCl3): δ 8.34 (s, 1H, (CO)H), 7.29 (s, 1H, 6’- 19.2 ppm.
HAr), 7.08 – 7.03 (m, 3H, 3’,4’,5’-HAr), 6.77 – 6.70 (m, 3H,
5, 6, 7-HAr), 5.75 (d, J = 9.1 Hz, 1H, N-H(CO)), 5.51 – 5.43 4.4.6 N-(6-iodo-2-(o-tolyl)-1,2,3,4-
(m, 1H, 4-Hax), 4.92 – 4.85 (m, 1H, 2-Hax), 3.76 (s, 1H, N- tetrahydroquinoline-4-yl)formamide (1f)
H), 1.33 (dd, J = 6.3, 3.8 Hz, 1H, 3b-Hec), 2.81 (hept, J = 6.7
Hz, 1H, 8-CH), 1.28 – 1.27 (m, 3H, 8-CH3), 1.27 – 1.26 (m, Dark green solid; yield: 0.28 g (0.71 mmol, 56%); mp:
3H, 8-CH3), 1.25 – 1.24 (m, 3H, 2’-CH3), 0.93 – 0.85 (m, 154,63 °C. IR (ATR): v max (cm-1) 3388, 3257, 3035, 2958,
1H, 3a-Hax) ppm. NMR 13C (101 MHz, CDCl3): δ 160.7, 2925, 2861, 1656, 1482, 1382, 1247, 1041, 756. NMR 1H
142.3, 140.7, 138.5, 137.7, 129.5, 128.6, 127.6, 127.1, 126.6, (400 MHz, CDCl3): δ 8.28 (s, 1H, (CO)H), 7.51 (d, J = 3.2
125.3, 124.5, 123.8, 50.9, 48.1, 38.7, 29.7, 24.8, 19.5 ppm. Hz, 1H, 6’-HAr), 7.43 (s, J = 1.6 Hz, 1H, 5-HAr), 7.33 (dd, J
= 8.5, 2.1 Hz, 1H, 7-HAr), 7.28 – 7.16 (m, 3H, 3’,4’,5’-HAr),
4.4.3 N-(6-methyl-2-(o-tolyl)-1,2,3,4- 6.37 (dd, J = 8.5, 2.1 Hz, 1H, 8-HAr), 5.88 (d, J = 9.3 Hz, 1H,
tetrahydroquinoline-4-yl)formamide (1c) N-H(CO)), 5.54 (ddd, J = 10.3, 10.0, 6.3 Hz, 1H, 4-Hax),
4.82 (dd, J = 10.8, 2.6 Hz, 1H, 2-Hax), 4.08 (s, 1H, N-H),
Beige solid; yield: 0.36 g (1.28 mmol, 71%); mp: 149.38 °C. 2.39 (s, 3H, 2’-CH3), 2.36 (d, J = 5.3 Hz, 1H, 3b-Hec), 1.90 –
IR (ATR): v max (cm-1) 3366, 3293, 3252, 3024, 2951, 2916, 1.82 (m, 1H, 3a-Hax) ppm. NMR 13C (101 MHz, CDCl3): δ
2851, 1667, 1615, 1506, 1233, 766. NMR 1H (400 MHz, 161.2, 145.1, 140.3, 137.2, 135.6, 135.2, 130.9, 127.7, 126.6,
CDCl3): δ 8.30 (s, 1H, (CO)H), 7.57 (d, J = 2.7 Hz, 1H, 6’- 125.5, 123.2, 116.7, 78.2, 51.6, 44.7, 36.4, 19.2 ppm.
HAr), 7.25 – 7.18 (m, 3H, 3’,4’,5’-HAr), 7.01 (s, 1H, 5-HAr),
6.93 (d, J = 8.2 Hz, 1H, 7-HAr), 6.53 (d, J = 8.1 Hz, 1H, 8- 4.4.7 N-(6-methoxy-2-(o-tolyl)-1,2,3,4-
HAr), 5.81 (d, J = 9.2 Hz, 1H, N-H(CO)), 5.58 (ddd, J = 10.0, tetrahydroquinoline-4-yl)formamide (1g)
9.9, 6.2 Hz, 1H, 4-Hax), 4.79 (dd, J = 10.9, 2.5 Hz, 1H, 2-
Hax), 3.90 (s, 1H, N-H), 2.44 (dd, J = 6.1, 2.5 Hz, 1H, 3b- Beige solid; yield: 0.39 g (1.32 mmol, 78%); mp: 151.56 °C.
Hec), 2.40 (s, 3H, 6-CH3), 2.26 (s, 3H, 2’-CH3), 1.91 – 1.80 IR (ATR): v max (cm-1) 3365, 3294, 3246, 3030, 2958, 2927,
(m, 1H, 3a-Hax) ppm. NMR 13C (101 MHz, CDCl3): δ 161.1, 2843, 1667, 1502, 1462, 1384, 1232, 1159, 1037, 809, 756,
143.2, 140.9, 135.2, 130.8, 129.3, 127.7, 127.5, 127.4, 126.6, 665. NMR 1H (400 MHz, CDCl3): δ 8.29 (s, 1H, (CO)H),
125.6, 120.7, 114.9, 51.9, 45.1, 37.2, 20.5, 19.2 ppm. 7.58 (d, J = 2.3 Hz, 1H, 6’-HAr), 7.25 – 7.19 (m, 3H, 3’,4’,5’-

https://doi.org/10.26434/chemrxiv-2023-7f4vv ORCID: https://orcid.org/0000-0003-0637-5885 Content not peer-reviewed by ChemRxiv. License: CC BY-NC-ND 4.0
9

HAr), 6.77 (s, 1H, 5-HAr), 6.73 (d, J = 8.6 Hz, 1H, 7-HAr), at a wavelength of 690 nm, obtaining a turbidity
6.56 (d, J = 8.6 Hz, 1H, 8-HAr), 5.86 (d, J = 9.3 Hz, 1H, N- (absorbance) of 0.082 corresponding to 0.5 on the
H(CO)), 5.60 (ddd, J = 10.1, 9.1, 6.3 Hz, 1H, 4-Hax), 4.76 McFarland scale, comparable to a bacterial suspension
(dd, J = 10.9, 2.4 Hz, 1H, 2-Hax), 3.76 (s, 1H, N-H), 3.75 (s, containing 1.5 x 108 CFU/mL [70]. After 12 hours, the
3H, 6-OCH3), 2.44 (dd, J = 6.2, 2.4 Hz, 1H, 3b-Hec), 2.40 (s, absorbance of the inoculum was measured to the 0.5
3H, 2’-CH3), 1.92 – 1.79 (m, 1H, 3a-Hax). NMR 13C (101 McFarland scale (absorbance approximately 0.082).
MHz, CDCl3): δ 161.12, 152.46, 140.86, 139.73, 135.16,
130.77, 127.48, 126.53, 125.61, 121.85, 116.03, 115.17, Antibacterial effect of N-(2-(o-tolyl)-1,2,3,4-
112.51, 77.40, 77.09, 76.77, 55.92, 52.07, 45.35, 37.24, tetrahydroquinoline-4-yl)formamide derivatives (1a-h)
19.16 ppm. on the bacterial culture of P. aeruginosa ATCC 27853
and E. coli 25922 (in situ). The assay was prepared
4.4.8 N-(5,8-dimethoxy-2-(o-tolyl)-1,2,3,4- according to the indications established by the Clinical &
tetrahydroquinoline-4-yl)formamide (1h) Laboratory Standards Institute (CLSI) [69]. 10 μL of a 0.5
McFarland culture were seeded in 200 μL Mueller-Hinton
White solid; yield: 0.19 g (0.58 mmol, 38%); mp: 126,25 °C. broth (MH) at various concentrations (10, 20, 35, 50, and 75
IR (ATR): v max (cm-1) 3267, 3198, 3135, 3067, 2970, 2873, μg/mL) of each of the synthetic autoinducers (1a-h) from a
1673, 1599, 1542, 1490, 1439, 1399, 1296, 1142, 748, 691. stock solution of 1000 μg/mL diluted in DMSO at 1%. This
NMR 1H (400 MHz, CDCl3): δ 7.78 (s, 1H, (CO)H), 7.39 (d, was done in corning 96-well plates and incubated at 37°C
J = 6.5 Hz, 1H, 6’-HAr), 7.19 – 7.16 (m, 3H, 3’,4’,5’-HAr), under agitation. Absorbance was measured at 690 nm for P.
6.20 (d, J = 3.2 Hz, 1H, 6-HAr), 6.17 (d, J = 3.3 Hz, 1H, 7- aeruginosa and 660 nm for E. coli every hour for 8 hours
HAr), 5.48 (q, J = 5.9 Hz, 1H, 4-Hax), 5.19 (d, J = 8.6 Hz, 1H, [69]. Each concentration of the compounds was evaluated in
N-H(CO)), 4.81 (d, J = 6.2 Hz, 1H, 2-Hax), 3.86 (s, 3H, 5- duplicate for an N=3, on the whole with the positive
OCH3), 3.77 (s, 1H, N-H), 3.76 (s, 3H, 8-OCH3), 2.50 – 2.45 inhibition control consisting of Gentamicin 10 μg/mL (C+),
(m, 1H, 3b-Hec), 2.39 (s, 3H, 2’-CH3), 2.34 – 2.22 (m, 1H, growth control (C-) consisting of bacterial culture without
3a-Hax). NMR 13C (101 MHz, CDCl3): δ 159.90, 152.81, compound, and cytotoxicity control being the bacterial
141.69, 140.61, 136.91, 135.00, 131.16, 127.15, 126.26, culture with DMSO at 1%. After the absorbance values were
125.05, 109.33, 107.91, 97.17, 56.03, 55.53, 49.90, 40.43, measured using the culture medium to calibrate, the
34.62, 19.09 ppm. percentages of inhibition in each measurement were
calculated using a T-test and one-way ANOVA, with the
4.5 Biological Assays GraphPad Prism 8.0 and Excel 2013 statistical package.

4.5.1 General procedure for the determination 4.5.2 General procedure for the identification of
of the antibacterial activity phenotypic switching on the bacterial
culture of P. aeruginosa ATCC 27853
Samples and reference control. The micro broth method
was used according to the protocol of the Clinical & Biofilm Formation Index. The tests were carried out on
Laboratory Standards Institute (CLSI) [69]. Stock solutions corning 96-well plates at the Laboratorio de Biología
were prepared for each compound, dissolving 1000 µg of Molecular e inmunología de la Universidad Libre seccional
each compound in 1 mL dimethyl sulfoxide (DMSO) 1%. Barranquilla. The 0.5 McFarland strain of P. aeruginosa was
The dilution equation was used to calculate the amount of sown in nutrient broth in the presence and absence of
compound to be used at each of the concentrations to be different concentrations (10, 20, 35, 50, and 75 μg/mL) of
tested in the bacterial culture (10, 20, 35, 50, and 75 μg/mL). tetrahydroquinoline compounds. Inoculated plates were
The different concentrations of the compounds were chosen incubated at 37°C for 24 hours. Then, the plates were gently
taking into account previous research where shaken in a microplate shaker to mix the planktonic cells.
tetrahydroquinolines were very effective against these The supernatant from each well was transferred to the
bacterial strains [39]. For the reference control, a stock corresponding well of a new microplate to measure the
solution of Gentamicin at 100 μg/mL was prepared from a absorbance at 600 nm (AbsRemaining). The remaining intact
concentrated solution (8 mg/mL). The amount of gentamicin biofilm in each well was stained with a 1% violet crystal
required for 2 mL culture strain and a final concentration of solution (CV) [71] at 37 °C for 30 min, then, washed with
10 μg/mL was determined. sterile distilled water and dried at 55 °C for 1 hour. Biofilm
cells were destained with 95% ethanol and absorbance was
Biological media. 0,013 g nutrient broth was added and measured at 570 nm (AbsBiofilm). The biofilm formation index
dissolved in 100 mL of ultra-pure water, then, it was was calculated with statistical significance using a T-test and
sterilized in an autoclave at 120°C with a pressure of 15 one-way ANOVA, with the GraphPad Prism 8.0 statistical
Pounds Per Square Inch (PSI) for approximately 45 minutes. package and Excel 2013.
In sterile test tubes, one inoculum of each bacterium was
seeded in 2 mL of sterile nutrient broth and incubated at 37 Pyocyanin Production. It was measured from the bacterial
°C for 12 hours [69]. The inoculum was standardized with a culture supernatant following the protocol reported by
0.5 McFarland pattern, which was prepared by adding 0.5 O’Loughlin et al. 2013 [70] with some modifications. 1 mL
mL BaCl2 0.048 M with 99.5 mL H2SO4 0.18 M and stirring of a 1:1000 subculture of P. aeruginosa was seeded in 9 mL
for 2 minutes. This solution was read in a spectrophotometer LB broth and incubated at 37°C. Then, a stock solution of

https://doi.org/10.26434/chemrxiv-2023-7f4vv ORCID: https://orcid.org/0000-0003-0637-5885 Content not peer-reviewed by ChemRxiv. License: CC BY-NC-ND 4.0
10

tetrahydroquinoline compounds (1a,c,d,f) was prepared in Supportive/Supplementary Material and include a brief
1% DMSO, and the corresponding quantities of each caption line for each file describing its contents.
compound were added in 1.5 mL microtubes to obtain
concentrations of 10, 20, 35, 50, and 75 μg/mL. The test Declaration of Generative AI and AI-assisted
inoculum was prepared by a 1:10 dilution of the 16-hour technologies in the writing process
culture in fresh medium and 200 μL aliquot of this
subculture was added to each of the microtubes and During the preparation of this work, the author(s) used
incubated at 37°C for approximately 20 hours. Then, Grammarly: Free Writing AI Assistance to check grammar,
absorbance was determined by spectrophotometric reading at style, and punctuation in the writing. After using this
600 nm. The remaining volume of each of the test tubes was tool/service, the author(s) reviewed and edited the content as
centrifuged at 2500 RPM at 15 °C for 10 minutes and 200 needed and take(s) full responsibility for the content of the
μL of the supernatant was transferred to new microplates for publication.
absorbance measurement at 695 nm.

5 Conclusion References

A versatile methodology was developed for the construction [1] O. Hernández-Gámez, O. Camacho-Romero, H. J.
of a serie of N-(2-(o-tolyl)-1,2,3,4-tetrahydroquinoline-4- González-Torres, S. Bolívar-González, M. Campo-
yl)formamide derivatives (1a-h) under simple reaction Urbina, and I. Zuluaga-De León, “Impact on the
conditions using p-TsOH as catalyst. In addition, bacterial resistance of the previous revision of the
antibacterial activity on bacterial strains of E. coli y P. prescription of antibiotics by the pharmaceutical
aeruginosa reported that compounds with electron- service in hospitals of Atlántico (Colombia),” Salud
withdrawing group substituents could be considered potent Uninorte, vol. 35, no. 2, pp. 187–204, 2019, doi:
growth inhibitors with inhibition percentages between 35- 10.14482/sun.35.2.615.1.
40%. On the other hand, the effectiveness of the THQ [2] D. V Hernández and A. . Hernández, “Resistencia a
compounds in the biofilm formation and pyocyanin los antibióticos, una amenaza latente,” Revista Aire
production assays on P. aeruginosa was evaluated to explore Libre, vol. 4, pp. 29–40, 2016, [Online]. Available:
the scope and biological limitations. Indeed, higher https://revia.areandina.edu.co/index.php/RAL/article
percentages of inhibition were found for the compounds N- /view/1014.
(6-fluor-2-(o-tolyl)-1,2,3,4-tetrahydroquinoline-4- [3] Samreen, I. Ahmad, H. A. Malak, and H. H.
yl)formamide 1a, N-(6-chloro-2-(o-tolyl)-1,2,3,4- Abulreesh, “Environmental antimicrobial resistance
tetrahydroquinoline-4-yl)formamide 1d, and N-(6-iodo-2-(o- and its drivers: a potential threat to public health,”
tolyl)-1,2,3,4-tetrahydroquinoline-4-yl)formamide 1f at Journal of Global Antimicrobial Resistance, vol. 27,
concentrations of 50 and 70 µg/mL, being N-(6-fluor-2-(o- pp. 101–111, 2021, doi: 10.1016/j.jgar.2021.08.001.
tolyl)-THQ derivative 1a the most effective agent in the [4] I. Samanta and S. Bandyopadhyay, “History of
serie. Consequently, the synthetic halogenated analogs antimicrobial resistance,” Antimicrobial Resistance
induced the bacterium P. aeruginosa to express moderate to in Agriculture, no. 1874, pp. 1–5, 2020, doi:
weak biofilm formation indices at the highest concentration 10.1016/b978-0-12-815770-1.00001-8.
evaluated, correlating with the low pyocyanin production at [5] M. Galindo-Méndez, “ Antimicrobial Resistance in
75 µg/mL. This suggests the biological activity of 1a-h Escherichia coli ,” E. Coli Infections - Importance of
compounds is led by QS modulation. Early Diagnosis and Efficient Treatment, 2020, doi:
10.5772/intechopen.93115.
Declaration of Competing Interest [6] A. Alarcon and F. Omenaca, “Antimicrobial
Resistance in Escherichia coli Sepsis,” Pediatric
The authors declare that they have no known competing Infectious Disease Journal, vol. 23, no. 10, pp. 979–
financial interests or personal relationships that could have 980, 2018, doi:
appeared to influence the work reported in this paper. 10.1097/01.inf.0000141751.73083.7a.
[7] D. Wu, Y. Ding, K. Yao, W. Gao, and Y. Wang,
Acknowledgments “Antimicrobial Resistance Analysis of Clinical
Escherichia coli Isolates in Neonatal Ward,”
L.J.G.M & D.F.O.F thank MINISTERIO and Fondo de Frontiers in Pediatrics, vol. 9, no. May, pp. 1–7,
Investigación en Salud (FIS, by its acronym in Spanish) 2021, doi: 10.3389/fped.2021.670470.
(CTeI;519-2021 from 874-2020) for the financial support; [8] P. K. Taylor, A. T. Y. Yeung, and R. E. W.
and Biomedicine Advanced Research Group (GIAB, by its Hancock, “Antibiotic resistance in Pseudomonas
acronym in Spanish). aeruginosa biofilms: Towards the development of
novel anti-biofilm therapies,” Journal of
Appendix A. Supplementary material Biotechnology, vol. 191, pp. 121–130, 2014, doi:
10.1016/j.jbiotec.2014.09.003.
Supportive/Supplementary material intended for publication [9] M. T. T. Thi, D. Wibowo, and B. H. A. Rehm,
must be numbered and referred to in the manuscript but “Pseudomonas aeruginosa biofilms,” International
should not be a part of the submitted paper. List all Journal of Molecular Sciences, vol. 21, no. 22, pp.

https://doi.org/10.26434/chemrxiv-2023-7f4vv ORCID: https://orcid.org/0000-0003-0637-5885 Content not peer-reviewed by ChemRxiv. License: CC BY-NC-ND 4.0
11

1–25, 2020, doi: 10.3390/ijms21228671. aeruginosa?,” Frontiers in Microbiology, vol. 10, no.
[10] S. M. Ribeiro et al., “New frontiers for anti-biofilm July, pp. 1–9, 2019, doi: 10.3389/fmicb.2019.01582.
drug development,” Pharmacology and [24] A. Ilangovan et al., “Structural Basis for Native
Therapeutics, vol. 160, pp. 133–144, 2016, doi: Agonist and Synthetic Inhibitor Recognition by the
10.1016/j.pharmthera.2016.02.006. Pseudomonas aeruginosa Quorum Sensing Regulator
[11] X. Bertrand and D. Hocquet, “Antibiotic drug PqsR (MvfR),” PLoS Pathogens, vol. 9, no. 7, 2013,
resistance: Causes and solutions,” EJHP Practice, doi: 10.1371/journal.ppat.1003508.
vol. 17, no. 2, pp. 58–59, 2011. [25] T. Yokoi, Y. Nakagawa, and H. Miyagawa,
[12] C. Ballot et al., “Essential role of mitochondria in “Asymmetric synthesis of tetrahydroquinoline-type
apoptosis of cancer cells induced by the marine ecdysone agonists and QSAR for their binding
alkaloid Lamellarin D,” Molecular Cancer affinity against Aedes albopictus ecdysone
Therapeutics, vol. 8, no. 12, pp. 3307–3317, 2009, receptors,” Pest Management Science, vol. 75, no. 1,
doi: 10.1158/1535-7163.MCT-09-0639. pp. 115–124, 2019, doi: 10.1002/ps.5160.
[13] Ventola CL, “The antibiotic resistance crisis: causes [26] V. V. Kouznetsov, C. M. Meléndez Gómez, M. G.
and threats.,” P & T journal, vol. 40, no. 4, pp. 277– Derita, L. Svetaz, E. Del Olmo, and S. A. Zacchino,
283, 2015. “Synthesis and antifungal activity of diverse C-2
[14] N. Høiby et al., “The clinical impact of bacterial pyridinyl and pyridinylvinyl substituted quinolines,”
biofilms,” International Journal of Oral Science, Bioorganic and Medicinal Chemistry, vol. 20, no.
vol. 3, no. 2, pp. 55–65, 2011, doi: 21, pp. 6506–6512, 2012, doi:
10.4248/IJOS11026. 10.1016/j.bmc.2012.08.036.
[15] O. Ciofu and T. Tolker-Nielsen, “Tolerance and [27] F. K. Behbahani and P. Ziaei, “Synthesis of 1,2,3,4-
resistance of pseudomonas aeruginosabiofilms to Tetrahydroquinolines using AlCl3 in aqua
antimicrobial agents-how P. aeruginosaCan escape mediated,” Journal of the Korean Chemical Society,
antibiotics,” Frontiers in Microbiology, vol. 10, no. vol. 58, no. 1, pp. 44–48, 2014, doi:
MAY, 2019, doi: 10.3389/fmicb.2019.00913. 10.5012/jkcs.2014.58.1.44.
[16] G. Rampioni, L. Leoni, and P. Williams, “The art of [28] A. Palanimuthu, C. Chen, and G. H. Lee,
antibacterial warfare: Deception through interference “Diastereoselective synthesis of highly substituted
with quorum sensing-mediated communication,” tetrahydroquinolines using benzoylacetonitrile via
Bioorganic Chemistry, vol. 55, pp. 60–68, 2014, doi: aza Diels–Alder reaction,” Tetrahedron Letters, vol.
10.1016/j.bioorg.2014.04.005. 72, p. 153063, 2021, doi:
[17] S. Singh, S. K. Singh, I. Chowdhury, and R. Singh, 10.1016/j.tetlet.2021.153063.
“Understanding the Mechanism of Bacterial [29] K. C. C. Aganda, B. Hong, and A. Lee, “Aerobic α-
Biofilms Resistance to Antimicrobial Agents,” The Oxidation of N-Substituted Tetrahydroisoquinolines
Open Microbiology Journal, vol. 11, no. 1, pp. 53– to Dihydroisoquinolones via Organo-
62, 2017, doi: 10.2174/1874285801711010053. photocatalysis,” Advanced Synthesis and Catalysis,
[18] D. Perez-Pascual, V. Monnet, and R. Gardan, vol. 361, no. 5, pp. 1124–1129, 2019, doi:
“Bacterial cell-cell communication in the host via 10.1002/adsc.201801301.
RRNPP peptide-binding regulators,” Frontiers in [30] Z. Zaheer, F. A. K. Khan, J. N. Sangshetti, R. H.
Microbiology, vol. 7, no. MAY, pp. 1–8, 2016, doi: Patil, and K. S. Lohar, “Novel amalgamation of
10.3389/fmicb.2016.00706. phthalazine–quinolines as biofilm inhibitors: One-
[19] S. Song and T. K. Wood, “The primary pot synthesis, biological evaluation and in silico
physiological roles of autoinducer 2 in escherichia ADME prediction with favorable metabolic fate,”
coli is chemotaxis and biofilm formation,” Bioorganic and Medicinal Chemistry Letters, vol.
Microorganisms, vol. 9, no. 2, pp. 1–6, 2021, doi: 26, no. 7, pp. 1696–1703, 2016, doi:
10.3390/microorganisms9020386. 10.1016/j.bmcl.2016.02.057.
[20] Y. Quan et al., “Regulation of bacteria population [31] I. Muthukrishnan, V. Sridharan, and J. C. Menéndez,
behaviors by AI-2 ‘consumer cells’ and ‘supplier “Progress in the Chemistry of
cells,’” BMC Microbiology, vol. 17, no. 1, pp. 1–9, Tetrahydroquinolines,” Chemical Reviews, vol. 119,
2017, doi: 10.1186/s12866-017-1107-2. no. 8, pp. 5057–5191, 2019, doi:
[21] S. Scutera, M. Zucca, and D. Savoia, “Novel 10.1021/acs.chemrev.8b00567.
approaches for the design and discovery of quorum- [32] S. L. Chua et al., “Reduced intracellular c-di-GMP
sensing inhibitors,” Expert Opinion on Drug content increases expression of quorum sensing-
Discovery, vol. 9, no. 4, pp. 353–366, 2014, doi: regulated genes in Pseudomonas aeruginosa,”
10.1517/17460441.2014.894974. Frontiers in Cellular and Infection Microbiology,
[22] M. F. Moradali, S. Ghods, and B. H. A. Rehm, vol. 7, no. OCT, pp. 1–8, 2017, doi:
“Pseudomonas aeruginosa lifestyle: A paradigm for 10.3389/fcimb.2017.00451.
adaptation, survival, and persistence,” Frontiers in [33] J. Liu et al., “Novel 2-Substituted 3-Hydroxy-1,6-
Cellular and Infection Microbiology, vol. 7, no. dimethylpyridin-4(1 H)-ones as Dual-Acting Biofilm
FEB, 2017, doi: 10.3389/fcimb.2017.00039. Inhibitors of Pseudomonas aeruginosa,” Journal of
[23] S. Yan and G. Wu, “Can Biofilm Be Reversed Medicinal Chemistry, vol. 63, no. 19, pp. 10921–
Through Quorum Sensing in Pseudomonas 10945, 2020, doi: 10.1021/acs.jmedchem.0c00763.

https://doi.org/10.26434/chemrxiv-2023-7f4vv ORCID: https://orcid.org/0000-0003-0637-5885 Content not peer-reviewed by ChemRxiv. License: CC BY-NC-ND 4.0
12

[34] I. Aleksić et al., “Long-Chain 4-Aminoquinolines as endo-fused 5-unsubstituted Hexahydro-2H-


Quorum Sensing Inhibitors in Serratia marcescens pyrano[3,2-c]quinolinesvia Sequential Sc(OTf) 3-
and Pseudomonas aeruginosa,” ACS Chemical catalyzed Cationic Imino-Diels-Alder Reaction/N-
Biology, vol. 12, no. 5, pp. 1425–1434, 2017, doi: debenzylation using N-benzylanilines, 3,4-dihydro-
10.1021/acschembio.6b01149. 2H-pyran and Paraformaldehyde under MW
[35] C. Mario, M. Gómez, and V. V Kouznetsov, “Small Irradiation,” Current organic synthesis, vol. 18, no.
molecules as regulators of bacterial quorum sensing. 5, pp. 431–442, Jan. 2021, doi:
New strategy in the development of antimicrobial 10.2174/1570179418666210113160949.
agents,” Antimicrobial Research: Novel [45] Y. B.R.D. Rajesh, “Quinoline Heterocycles:
Bioknowledge and Educational Programs, pp. 610– Synthesis and Bioactivity,” Heterocycles - Synthesis
622, 2017. and Biological Activities, 2020, doi:
[36] E. T. Anthony, M. O. Ojemaye, O. O. Okoh, and A. 10.5772/intechopen.81239.
I. Okoh, “A critical review on the occurrence of [46] R. Shimizu, Y. Okada, and K. Chiba, “Stepwise
resistomes in the environment and their removal radical cation Diels-Alder reaction via multiple
from wastewater using apposite treatment pathways,” Beilstein Journal of Organic Chemistry,
technologies: Limitations, successes and future vol. 14, pp. 704–708, 2018, doi: 10.3762/bjoc.14.59.
improvement,” Environmental Pollution, vol. 263, p. [47] D. Wei, A. Bruneau-Voisine, D. A. Valyaev, N.
113791, 2020, doi: 10.1016/j.envpol.2019.113791. Lugan, and J. B. Sortais, “Manganese catalyzed
[37] C. A. Michael, D. Dominey-Howes, and M. Labbate, reductive amination of aldehydes using hydrogen as
“The antimicrobial resistance crisis: Causes, a reductant,” Chemical Communications, vol. 54, no.
consequences, and management,” Frontiers in 34, pp. 4302–4305, 2018, doi: 10.1039/c8cc01787e.
Public Health, vol. 2, no. SEP, pp. 1–8, 2014, doi: [48] G. Li, G. Yu, C. Wang, T. Morita, X. Zhang, and H.
10.3389/fpubh.2014.00145. Nakamura, “Supporting Information Synthesis of 2-
[38] M. D. Matveeva, R. Purgatorio, L. G. Phosphonotetrahydroquinolines with Site-Selective
Voskressensky, and C. D. Altomare, “Pyrrolo[2,1- C-H bond Phosphonylation through I ntramolecular
a]isoquinoline scaffold in drug discovery: Advances Hydroarylation-Redox CDC Reaction,” 2021.
in synthesis and medicinal chemistry,” Future [49] I. Muthukrishnan et al., “Heterogeneous Amberlyst-
Medicinal Chemistry, vol. 11, no. 20, pp. 2735– 15-catalyzed synthesis of complex hybrid
2755, 2019, doi: 10.4155/fmc-2019-0136. heterocycles containing [1,6]-naphthyridine under
[39] H. M. Faidallah, A. A. Saqer, K. A. Alamry, K. A. metal-free green conditions,” Organic and
Khan, and A. M. Asiri, “Synthesis and biological Biomolecular Chemistry, vol. 17, no. 28, pp. 6872–
evaluation of some novel tetrahydroquinolines as 6879, 2019, doi: 10.1039/c9ob01256g.
anticancer and antimicrobial agents,” Journal of [50] J. Pasha et al., “Amberlyst-15 catalyzed Povarov
Enzyme Inhibition and Medicinal Chemistry, vol. 29, reaction of N-arylidene-1H-indazol-6-amines and
no. 3, pp. 367–378, 2014, doi: indoles: A greener approach to the synthesis of exo-
10.3109/14756366.2013.787421. 1,6,7,7a,12,12a-hexahydroindolo[3,2-c]pyrazolo[3,4-
[40] P. N. Chavan, D. N. Pansare, S. L. Jadhav, and M. J. f]quinolines as potential sirtuin inhibitors,”
Rai, “Synthesis and biological activities of new Tetrahedron Letters, vol. 56, no. 18, pp. 2289–2292,
tetrahydroquinoline and pyrimidine derivatives,” 2015, doi: 10.1016/j.tetlet.2015.03.078.
European Chemical Bulletin, vol. 8, no. 8, pp. 257– [51] M. Gutiérrez, U. Carmona, G. Vallejos, and L.
264, 2019, doi: 10.17628/ecb.2019.8.257-264. Astudillo, “Antifungal activity of
[41] J. Liu et al., “Design, synthesis, and biological tetrahydroquinolines against some phytopathogenic
evaluation of tetrahydroquinoline amphiphiles as fungi,” Zeitschrift fur Naturforschung - Section C
membrane-targeting antimicrobials against Journal of Biosciences, vol. 67 C, no. 11–12, pp.
pathogenic bacteria and fungi,” European Journal of 551–556, 2012, doi: 10.1515/znc-2012-11-1204.
Medicinal Chemistry, vol. 243, no. May, p. 114734, [52] M. T. Madigan, J. M. Martinko, K. S. Bender, D. H.
2022, doi: 10.1016/j.ejmech.2022.114734. Buckley, and D. A. Stahl, Brock. Biología de los
[42] C. M. Meléndez Gómez, M. Marsiglia, R. Escarsena, microorganismos, vol. 14, no. 1. 2015.
E. del Olmo, and V. V. Kouznetsov, “Synthesis of [53] C. D. M. Wijers, L. Pham, M. V Douglass, E. P.
2,3-di(ω-hydroxyalkyl)quinolines from anilines and Skaar, L. D. Palmer, and M. J. Noto, “Gram-negative
cyclic enols using sequential bacteria act as a reservoir for aminoglycoside
cycloaddition/aromatization reactions,” Tetrahedron antibiotics that interact with host factors to enhance
Letters, vol. 59, no. 1, pp. 22–25, 2018, doi: bacterial killing in a mouse model of pneumonia,”
10.1016/j.tetlet.2017.11.034. FEMS Microbes, vol. 3, no. May, pp. 1–13, 2022,
[43] R. A. Mekheimer, M. A. Al-Sheikh, H. Y. Medrasi, doi: 10.1093/femsmc/xtac016.
and K. U. Sadek, “Advancements in the synthesis of [54] M. Esther, T. Alarcón, N. P. Pájaro, and G. L.
fused tetracyclic quinoline derivatives,” RSC Méndez, “esenciales de diferentes especies del
Advances, vol. 10, no. 34, pp. 19867–19935, 2020, género Citrus Resumen Antibacterial activity in vitro
doi: 10.1039/d0ra02786c. of essential oils Introducción,” vol. 46, no. 2, pp.
[44] A. R. M. Salgado, C. E. P. Galvis, V. V. 160–175, 2017.
Kouznetsov, and C. M. Meléndez, “Synthesis of [55] M. Alagumuthu and S. Arumugam, “Molecular

https://doi.org/10.26434/chemrxiv-2023-7f4vv ORCID: https://orcid.org/0000-0003-0637-5885 Content not peer-reviewed by ChemRxiv. License: CC BY-NC-ND 4.0
13

docking, discovery, synthesis, and pharmacological role of pyocyanin in Pseudomonas aeruginosa


properties of new 6-substituted-2-(3- infection,” Trends in Molecular Medicine, vol. 10,
phenoxyphenyl)-4-phenyl quinoline derivatives; an no. 12, pp. 599–606, 2004, doi:
approach to developing potent DNA gyrase 10.1016/j.molmed.2004.10.002.
inhibitors/antibacterial agents,” Bioorganic and [67] S. J. Ho Sui et al., “Raloxifene attenuates
Medicinal Chemistry, vol. 25, no. 4, pp. 1448–1455, Pseudomonas aeruginosa pyocyanin production and
2017, doi: 10.1016/j.bmc.2017.01.007. virulence,” International Journal of Antimicrobial
[56] T. D. M. Pham, Z. M. Ziora, and M. A. T. Agents, vol. 40, no. 3, pp. 246–251, 2012, doi:
Blaskovich, “Quinolone antibiotics,” 10.1016/j.ijantimicag.2012.05.009.
MedChemComm, vol. 10, no. 10, pp. 1719–1739, [68] C. T. O’Loughlin, L. C. Miller, A. Siryaporn, K.
2019, doi: 10.1039/c9md00120d. Drescher, M. F. Semmelhack, and B. L. Bassler, “A
[57] A. Dorababu, “Recent update on antibacterial and quorum-sensing inhibitor blocks Pseudomonas
antifungal activity of quinoline scaffolds,” Archiv aeruginosa virulence and biofilm formation,”
der Pharmazie, vol. 354, no. 3, pp. 1–20, 2021, doi: Proceedings of the National Academy of Sciences of
10.1002/ardp.202000232. the United States of America, vol. 110, no. 44, pp.
[58] D. Jiang, “4-Quinolone Derivatives and Their 17981–17986, 2013, doi: 10.1073/pnas.1316981110.
Activities Against Gram-negative Pathogens,” [69] N. Dayal et al., “Inhibitors of Intracellular Gram-
Journal of Heterocyclic Chemistry, vol. 55, no. 9, Positive Bacterial Growth Synthesized via Povarov-
pp. 2003–2018, 2018, doi: 10.1002/jhet.3244. Doebner Reactions,” ACS Infectious Diseases, vol.
[59] L. Senerovic, D. Opsenica, I. Moric, I. Aleksic, M. 5, no. 11, pp. 1820–1830, 2019, doi:
Spasić, and B. Vasiljevic, “Quinolines and 10.1021/acsinfecdis.9b00022.
quinolones as antibacterial, antifungal, anti- [70] G. E. Lozano, S. R. Beatriz, F. M. Cervantes, G. N.
virulence, antiviral and anti-parasitic agents,” P. María, and J. M. C. Francisco, “Low accuracy of
Advances in Experimental Medicine and Biology, the McFarland method for estimation of bacterial
vol. 1282, no. September 2019, pp. 37–69, 2020, doi: populations,” African Journal of Microbiology
10.1007/5584_2019_428. Research, vol. 12, no. 31, pp. 736–740, 2018, doi:
[60] E. Önem, B. Tüzün, and S. Akkoç, “Anti-quorum 10.5897/ajmr2018.8893.
sensing activity in Pseudomonas aeruginosa PA01 of [71] Q. Guo et al., “Potential use of dimethyl sulfoxide in
benzimidazolium salts: electronic, spectral and treatment of infections caused by Pseudomonas
structural investigations as theoretical approach,” aeruginosa,” Antimicrobial Agents and
Journal of Biomolecular Structure and Dynamics, Chemotherapy, vol. 60, no. 12, pp. 7159–7169,
vol. 40, no. 15, pp. 6845–6856, 2022, doi: 2016, doi: 10.1128/AAC.01357-16.
10.1080/07391102.2021.1890222.
[61] M. P. Storz et al., “Validation of PqsD as an anti-
biofilm target in pseudomonas aeruginosa by
development of small-molecule inhibitors,” Journal
of the American Chemical Society, vol. 134, no. 39,
pp. 16143–16146, 2012, doi: 10.1021/ja3072397.
[62] F. Casilag, A. Lorenz, J. Krueger, F. Klawonn, S.
Weiss, and S. Häussler, “LasB elastase of
Pseudomonas aeruginosa acts in concert with
alkaline protease AprA to prevent flagellin-mediated
immune recognition,” Infection and Immunity, vol.
84, no. 1, pp. 162–171, 2015, doi:
10.1128/IAI.00939-15.
[63] P. L. Faleiro Naves, Formación de biopelículas por
Escherichia coli y su correlación con factores de
virulencia : prevención y actividad de
antimicrobianos frente a organismos planctónicos y
asociados a biopelículas. 2010.
[64] B. Rada et al., “Pyocyanin-Enhanced Neutrophil
Extracellular Trap Formation Requires the NADPH
Oxidase,” PLoS ONE, vol. 8, no. 1, 2013, doi:
10.1371/journal.pone.0054205.
[65] C. S. Curran, T. Bolig, and P. Torabi-Parizi,
“Mechanisms and targeted therapies for
pseudomonas aeruginosa lung infection,” American
Journal of Respiratory and Critical Care Medicine,
vol. 197, no. 6, pp. 708–727, 2018, doi:
10.1164/rccm.201705-1043SO.
[66] G. W. Lau, D. J. Hassett, H. Ran, and F. Kong, “The

https://doi.org/10.26434/chemrxiv-2023-7f4vv ORCID: https://orcid.org/0000-0003-0637-5885 Content not peer-reviewed by ChemRxiv. License: CC BY-NC-ND 4.0

You might also like