1. Introduction
Helicobacter pylori, a pathogenic member of phylum
Campylobacterota (formerly
Epsilonproteobacteria) [
1], is recognized as the leading cause of several human gastric pathologies, including acute and chronic gastritis, peptic ulcer disease, gastric adenocarcinoma, and gastric mucosa-associated lymphoid tissue (MALT) lymphoma [
2,
3]. In addition, this microaerophilic Gram-negative bacterium has been associated to some extragastric disorders such as idiopathic thrombocytopenic purpura, idiopathic iron deficiency anaemia, vitamin B
12 deficiency, cardiovascular and neurodegenerative diseases, among others [
4].
H. pylori is presumably colonizing the gastric mucosa of more than half of the world population [
5], causing chronic long-lasting inflammation that may progress to gastric cancer in about 1-3% of untreated or mistreated cases [
6,
7]. Despite
H. pylori constitutes the only bacterial pathogen recognized as class I carcinogen by the International Agency for Research on Cancer [
8], neither prophylactic nor therapeutic vaccines are currently available.
Effective treatment of
H. pylori infection remains a challenge for clinicians and sanitary authorities worldwide [
9,
10,
11]. Only a handful of antibiotics are currently used in clinical practice for
H. pylori eradication, including metronidazole, clarithromycin, amoxicillin, tetracycline, and levofloxacin [
12,
13]. Due to the poor
in vivo efficacy of antimicrobial monotherapies against this pathogen [
14], current antibiotics are administrated as part of several aggressive and prolonged combinatory therapies which are commonly prescribed empirically, resulting frequently in
H. pylori eradication failures, emergence of secondary antibiotic resistances, and refractory infections [
15,
16,
17,
18]. In the last two decades, the alarming increase of the antibiotic resistance levels to first-line and even “rescue” antibiotics, especially clarithromycin, metronidazole and levofloxacin [
19,
20,
21], has led to a marked decrease of the eradication rates of traditional therapies [
22,
23]. This fact prompted the World Health Organization (WHO) to define
H. pylori as a high priority pathogen in the current global efforts to R&D of novel antimicrobials [
24].
On the other hand, even when current antimicrobial therapies against
H. pylori infections are generally safe, the prolonged administration of high doses of broad-spectrum antibiotics into combinatory therapies frequently result in dysbiosis [
25,
26,
27,
28]. In some cases,
H. pylori eradication regimens might cause major disruption of normal gut microbiome, promoting severe complications such as pseudomembranous colitis due to
Clostridioides difficile proliferation [
29,
30,
31,
32].
The discovery and development of narrow-spectrum antibiotics, also known as precision antimicrobials, exemplify a current accurate strategy for reducing or slowing down the appearance and propagation of antimicrobial resistance as well as minimizing undesirable side effects on normal microbiome [
33,
34,
35,
36,
37]. Ideally, a precision antimicrobial exerts its action on molecular targets shared by only a reduced number of closely related pathogenic species, but not expressed by most representative commensal microbes [
38,
39].
In previous works, we have validated the essential protein HsrA as an effective therapeutic target for
H. pylori infection [
40,
41,
42]. HsrA is an OmpR/PhoB-type orphan response regulator [
43,
44], unique and highly conserved in members of phylum
Campylobacterota [
45], which appears involved in a variety of crucial physiological processes including transcription, translation, redox homeostasis, chemotaxis and energy metabolism [
46,
47,
48,
49], and thereby resulting indispensable for cell viability [
50,
51]. In the present study, we have identified several novel low-molecular weight inhibitors of HsrA, which fit the Lipinski´s rule-of-five for oral drug-likeness and exhibited potent bactericidal activities against
H. pylori, with minor antimicrobial effects on both Gram-negative and Gram-positive species of commensal bacteria. At least one of these novel precision antimicrobials exerted also a strong antimicrobial action against
Campylobacter jejuni, another clinically relevant pathogen of phylum
Campylobacterota.
3. Discussion
Infection with
H. pylori constitutes a silent pandemic of global concern. About 50% of the world´s population is estimated to be infected with this carcinogenic bacterial pathogen [
5,
62], although this prevalence reaches 80% or more in several countries of Americas, Eastern Mediterranean, and Eastern Europe [
63].
H. pylori is considered the most important risk factor for development of gastric cancer [
7], the fifth most common malignancy and the fourth leading cause of all cancer-related deaths worldwide [
64]. Despite eradication of
H. pylori infection significantly reduces both incidence and mortality of gastric cancer [
65,
66,
67], the increasing resistome accumulated by this pathogen in the last years has led to a considerable drop in the efficacy of current eradication therapies [
10,
68,
69]. To face this concerning trend, efforts have being made to discover novel treatment strategies.
We previously validated the essential OmpR-like orphan response regulator HsrA as an effective therapeutic target against
H. pylori [
40,
41,
42]. HsrA affinity-based high-throughput screening (HTS) of 1,200 clinically approved drugs from Prestwick Chemical Library® revealed that some highly prescribed 1,4-dihydropyridine (DHP) calcium channel blockers, as well as several natural flavonoids, acted as HsrA low-molecular weight ligands leading to inhibition of the
in vitro DNA-binding activity of the regulator. Some of these potentially repurposable drugs exhibited noticeable bactericidal effects against different antibiotic-resistant strains of
H. pylori with MIC ranging 4 - 16 mg/L [
40,
41], and significantly decreased gastric colonization of
H. pylori in mice [
41]. Notably, flavonoids such as chrysin and hesperetin demonstrated strong synergistic bactericidal action against
H. pylori in combination with clarithromycin or metronidazole, supporting the potential inclusion of these natural phytochemicals as valuable adjuvants in novel eradication therapies [
40,
70].
In the present work, we have identified novel low-molecular weight inhibitors of HsrA through the screening of 12,000 drug-like compounds from the Maybridge HitFinder
TM chemical library. The collection is highly compliant with the Lipinski's rule-of-five, a rule of thumb to evaluate drug-likeness of novel potential medicines [
52,
53]. According to the Lipinski's rule-of-five, those compounds with molecular weight < 500 Da, calculated logarithm of the octanol-water partition coefficient (clog
P) < 5, number of hydrogen bond acceptors ≤ 10, and number of hydrogen bond donors ≤ 5, will have better oral bioavailability and pharmacokinetics. Thus, molecules that violate more than one of these physicochemical parameters may show problems in terms of bioavailability after oral administration.
At least six of the novel drug-like HsrA ligands identified by affinity-based HTS of the HitFinder
TM collection, denoted as I, IV, V, VIII, XI and XII, noticeably inhibited the DNA binding activity of the response regulator
in vitro and demonstrated strong bactericidal activities against antibiotic-resistant strains of
H. pylori. With MIC values ranging from 0.031 to 4 mg/L, some of these novel antimicrobial candidates constituted the most potent bactericidal HsrA inhibitors discovered to date. In addition, at least four of these highly bactericidal HsrA ligands exhibited narrow-spectrum antimicrobial activities against
H. pylori, with MBC values ≥ 64 mg/L against several representative Gram-positive and Gram-negative members of normal human microbiota. Since protein HsrA is unique among members of phylum
Campylobacterota (former class
Epsilonproteobacteria) [
45], the results suggest that certain low-molecular weight inhibitors of HsrA could specifically bind this response regulator without affecting biological activities of essential proteins from other bacterial species. These evidences push up the value of HsrA as an effective and selective therapeutic target for development of novel precision antibiotics against
H. pylori, moving away the risk of dysbiosis observed with current eradication therapies [
28,
31].
Notably, one of the novel drug-like HsrA inhibitor described here, denoted as ligand V and named N'-{[(4-chloroanilino)carbonyl]}oxy-5-nitrothiophene-3-carboximidamide at the ChemSpider database (
https://www.chemspider.com), showed significantly higher antimicrobial activities than the rest of compounds against all
H. pylori strains used in the study. With MIC values ranging from 0.031 to 0.125 mg/L, the antimicrobial potency of ligand V was at least 4-fold higher than the second most active bactericidal HsrA inhibitor, the ligand XII, and more than 10-fold stronger than metronidazole against the same strains. In addition, ligand V demonstrated the highest therapeutic index among all highly bactericidal HsrA inhibitors described here, and showed additive antimicrobial action in combination with clarithromycin and metronidazole.
Several factors could be contributing to the higher antimicrobial potency exhibited by ligand V on
H. pylori cells. For instance, a favourable balance among the physicochemical properties of this compound could make a difference in the microbial membrane translocation and intracellular concentration compared with other HsrA bactericidal ligands. In this context, the lipophilicity/hydrophilicity ratio, the molecular weight, the structural flexibility (rotatable bonds), the number of H-bond donors and acceptors, but also the polar surface area of small molecules may greatly influence their permeability through bacterial membranes as well as their possible efflux outside the pathogen [
71,
72,
73,
74]. Despite the contribution of each physicochemical property to membrane permeation and efflux varies among different bacteria species [
74], it is well recognized that the polar nature of the outer membrane of Gram-negative pathogens hinders passive translocation of highly hydrophobic molecules [
71,
75]. Thus, decreasing the lipophilicity/hydrophilicity ratio of a low-molecular weight inhibitor of
E.coli DNA gyrase by reducing logD from 2.59 to 1.75 led to 4-fold increase in its antibacterial potency [
72]. Likewise, fine-tuning the physicochemical properties of a topoisomerase inhibitor in
Pseudomonas aeruginosa by lowering logD from 2.0 to 0.9 resulted in 8-fold increase of its antibacterial activity [
75]. On the other hand, Gram-negative pathogens express a plethora of broad-specific efflux pumps that actively export a variety of foreign molecules out of the cell, thereby decreasing the effective antibiotic concentration in the cytoplasm [
76]. Some evidences suggested that highly polar and small compounds or very large and zwitterionic molecules are less susceptible to efflux [
71].
Herein, time-kill kinetic studies demonstrated that ligand V and VIII produced a significant faster decline in bacterial counts compared to the rest of HsrA bactericidal inhibitors, suggesting similar permeability through
H. pylori membrane. However, the polar surface area (TPSA) of ligand V almost duplicates this property in ligand VIII, which could be linked with a higher propensity of the last compound to be expelled by
H. pylori efflux pumps. In fact, almost 30 genes related to different efflux pumps have been identified in the
H. pylori genome [
77].
The antimicrobial potency of HsrA ligands could also be influenced by the binding affinity of each molecule to its target protein. According to our ITC studies, the interaction of ligand V with HsrA occurs with > 3-fold higher affinity than the interaction of ligand XII, the second most potent bactericidal compound. Despite ligand IV bound HsrA with the highest affinity, its larger molecular weight and lipophilicity could negatively affect membrane permeation.
Besides its potent antimicrobial activity against
H. pylori, ligand V stood up for its strong bactericidal effect against
C. jejuni, another clinically relevant pathogen of phylum
Campylobacterota.
C. jejuni is considered the leading cause of food-borne bacterial gastroenteritis worldwide [
78]. Infection in humans usually occurs by consumption of contaminated foods (mainly poultry) or direct contact with animal hosts (including pets) and environmental reservoirs [
79,
80]. Despite most cases of human campylobacteriosis produces mild and self-limiting diarrhoea and may not require antimicrobial therapy [
78,
81], the use of antibiotics could be necessary to shorten the duration of illness and to prevent severe complications in more susceptible populations, including very young children, elderly people and immunocompromised patients [
78,
82]. In addition, antibiotics must be used in cases of invasive or extra-gastrointestinal manifestations such as meningitis, bacteraemia and endocarditis, among others [
81,
83,
84].
When antibiotics are needed, the recommended first-line treatment of campylobacteriosis consists of macrolides such as azithromycin and erythromycin [
85]. Fluoroquinolones like ciprofloxacin can be used as alternative therapy, though intravenous aminoglycosides or carbapenems are recommended in severe systemic cases. Tetracyclines and chloramphenicol are also alternative therapies, but they are usually avoided in young children due to their adverse effects [
82,
85]. As in the case of
H. pylori, the increasing rates of antibiotic resistance in
Campylobacter strains isolated from humans and animals worldwide, especially to fluoroquinolones but also to macrolides [
86,
87,
88], prompted the WHO to include
C. jejuni as a high priority pathogen in the search of novel antimicrobials [
24].
Among all the novel drug-like HsrA bactericidal inhibitors described in this work, only the ligand V exhibited a strong antimicrobial effect on
C. jejuni, with MIC = 0.25 mg/L. This antimicrobial potency is similar to that observed with ciprofloxacin, and stronger than those observed with doxycycline, erythromycin and gentamicin against the same
C. jejuni strain used herein [
89]. Molecular interaction of ligand V with CosR was demonstrated by ITC analysis, while this compound noticeable inhibited the
in vitro DNA binding activity of CosR according to EMSA experiments. Hence, the strong antimicrobial activity of ligand V on
C. jejuni appears to rely on the inhibition of the essential biological activity of CosR [
90,
91,
92,
93], thereby validating this protein as an effective therapeutic target against
C. jejuni [
44]. The dual inhibition triggered by the same low-molecular weight ligand on the biological activities of HsrA and CosR could be expected given the high sequence identity of both orthologue proteins, especially in their C-terminal DNA binding domains (60% overall identity, 85% identity in the effector domain) [
45].
Transcriptomic analysis was carried out in order to discern the global effects of lethal concentrations of ligand V on the
H. pylori physiology. The results observed here appeared in correspondence with an effective
in vivo inhibition of HsrA, and confirmed previous experimental evidences of Pelliciari and colleagues [
49] about the key role of this essential response regulator in the control of protein biosynthesis and energy metabolism. Exposure of
H. pylori to the highly bactericidal HsrA inhibitor V particularly disturbed transcription of genes involved in ribosome biogenesis [
94]. The strong detriment observed in the expression of a plethora of ribosomal proteins could certainly arrest translation and protein biosynthesis leading to cell death [
95]. In addition, the
in vivo inhibition of HsrA led to a significant decrease in the transcription of several components of the respiratory chain and ATP synthase. Proteome imbalance in the electron transport chain could impair redox reactions and proton translocation across cytoplasmic membrane, shrinking the transmembrane electrochemical proton gradient that drives the synthesis of ATP [
96,
97]. Furthermore, disruption of ATP biosynthesis could be additionally exacerbated by an altered expression of ATP synthase subunits. On the other hand, imbalance in the respiratory chain may increase the production of reactive oxygen species leading to oxidative stress [
98]. This deleterious condition could induce the expression of antioxidant enzymes, heat shock proteins, and other molecular chaperones [
99,
100], a transcriptional response observed in this work upon exposure of
H. pylori to the HsrA inhibitor V. Taking together, the bactericidal effect triggered by the
in vivo inhibition of the essential regulatory role of HsrA seems to be a consequence of synergistic deleterious disturbances in the
H. pylori physiology, including the arrest of protein biosynthesis, malfunction of respiratory chain, detriment in ATP generation, and oxidative stress.
Due to the unsuccessful attempts to manipulate the expression of the essential
hsrA gene
in vivo [
45,
47], the elucidation of the HsrA regulon has represented a challenge. Previous studies have identified several dozens of putative target genes and a handful of well-characterized targets [
46,
47,
48,
49]. Our transcriptomic data suggest that HsrA acts as a global transcriptional regulator, modulating directly or indirectly the expression of up to 23% of the ORFs encoded by the
H. pylori 26696 genome. Further analyses must be conducted in order to completely unravel the global regulatory role of HsrA and its crucial contribution to viability and pathogenicity of
H. pylori.
In conclusion, the results presented here strongly support the use of HsrA and its orthologue proteins as effective and selective therapeutic targets for development of novel antimicrobial strategies against clinically relevant pathogenic members of phylum Campylobacterota. The novel drug-like highly bactericidal inhibitors of HsrA described here constitute valuable candidates to a new family of narrow-spectrum antibiotics that allow overcoming the current resistome, protecting from dysbiosis, and enhancing the battery of therapeutic options for novel personalized treatments against H. pylori.
Author Contributions
Conceptualization, A.G.; methodology, A.G., J.C., I.O.-M., S.A., E.C., S.S. and A.V.-C.; software, A.G., J.C., I.O-M., and A.V.-C.; validation, A.G., J.C., I.O.-M., S.S., A.V.-C., E.P., M.F. and J.S.; formal analysis, A.G., I.O.-M., A.V.-C., E.P., M.F. and J.S ; investigation, A.G., J.C., I.O.-M., S.A., E.C., S.S. and A.V.-C.; resources, S.A., M.F., and A.L.; data curation, A.G., J.C., I.O.-M., S.A., E.C., S.S. and A.V.-C.; writing—original draft preparation, A.G.; writing—review and editing, A.G., J.C., I.O.-M., M.F., J.S. and A.L.; visualization, A.G., J.C., I.O.-M. and A.V.-C.; supervision, A.G., E.P., M.F., J.S., A.L.; project administration, A.G. and A.L.; funding acquisition, M.F. and A.L. All authors have read and agreed to the published version of the manuscript..
Figure 1.
Upshifts in the thermal unfolding curve of response regulator HsrA triggered by ligands IV and XII. The high-throughput screening (HTS) of the Maybridge HitFinderTM chemical library for HsrA ligands was assessed by a fluorescence-based thermal shift assay. Mixtures of DMSO (vehicle) instead of compounds were used as reference controls. Any compound that preferentially binds to the native state of HsrA would increase protein stability and cause an increase of the midpoint temperature of unfolding (Tm). Those compounds that clearly increased the thermal stability of HsrA observed in reference controls (Tm ≥ 5ºC) were considered as HsrA ligands.
Figure 1.
Upshifts in the thermal unfolding curve of response regulator HsrA triggered by ligands IV and XII. The high-throughput screening (HTS) of the Maybridge HitFinderTM chemical library for HsrA ligands was assessed by a fluorescence-based thermal shift assay. Mixtures of DMSO (vehicle) instead of compounds were used as reference controls. Any compound that preferentially binds to the native state of HsrA would increase protein stability and cause an increase of the midpoint temperature of unfolding (Tm). Those compounds that clearly increased the thermal stability of HsrA observed in reference controls (Tm ≥ 5ºC) were considered as HsrA ligands.
Figure 2.
Specific in vitro inhibition of the DNA binding activity of response regulator HsrA by selected ligands, according to EMSA. Recombinant HsrA was mixed with its target promoter PporG in the presence of 2, 1, 0.5, and 0.1 mM of ligands. DMSO instead of inhibitors was included as vehicle controls, while an internal sequence of the Anabaena sp. pkn22 gene was used as non-specific competitor in all assays. Addition of protein in the reaction mixtures are indicate by +. Protein-DNA interactions were analysed by 6% non-denaturing electrophoresis.
Figure 2.
Specific in vitro inhibition of the DNA binding activity of response regulator HsrA by selected ligands, according to EMSA. Recombinant HsrA was mixed with its target promoter PporG in the presence of 2, 1, 0.5, and 0.1 mM of ligands. DMSO instead of inhibitors was included as vehicle controls, while an internal sequence of the Anabaena sp. pkn22 gene was used as non-specific competitor in all assays. Addition of protein in the reaction mixtures are indicate by +. Protein-DNA interactions were analysed by 6% non-denaturing electrophoresis.
Figure 3.
Time-kill kinetics of selected HsrA inhibitors against Helicobacter pylori strain ATCC 700392. Fresh grown cells of H. pylori (104 CFU/mL) were exposed to 4 × MIC of each compound. Mixtures of bacteria with DMSO (vehicle) instead of inhibitors were used as controls. CFU were determined at time zero and after 2, 4, 8, and 24 h of exposure. Values are the means of three independent determinations; standard deviations are represented by vertical bars.
Figure 3.
Time-kill kinetics of selected HsrA inhibitors against Helicobacter pylori strain ATCC 700392. Fresh grown cells of H. pylori (104 CFU/mL) were exposed to 4 × MIC of each compound. Mixtures of bacteria with DMSO (vehicle) instead of inhibitors were used as controls. CFU were determined at time zero and after 2, 4, 8, and 24 h of exposure. Values are the means of three independent determinations; standard deviations are represented by vertical bars.
Figure 4.
The HsrA ligand V noticeably inhibited the in vitro DNA binding activity of the C. jejuni response regulator CosR, an orthologue protein of HsrA. Recombinant CosR was mixed with its target promoter PsodB in the presence of 2, 1, 0.5, and 0.1 mM of ligand V. DMSO instead of inhibitors was included as vehicle controls, while an internal sequence of the Anabaena sp. pkn22 gene was used as non-specific competitor in all assays. Addition of protein in the reaction mixtures are indicate by +. Protein-DNA interactions were analysed by 6% non-denaturing electrophoresis.
Figure 4.
The HsrA ligand V noticeably inhibited the in vitro DNA binding activity of the C. jejuni response regulator CosR, an orthologue protein of HsrA. Recombinant CosR was mixed with its target promoter PsodB in the presence of 2, 1, 0.5, and 0.1 mM of ligand V. DMSO instead of inhibitors was included as vehicle controls, while an internal sequence of the Anabaena sp. pkn22 gene was used as non-specific competitor in all assays. Addition of protein in the reaction mixtures are indicate by +. Protein-DNA interactions were analysed by 6% non-denaturing electrophoresis.
Figure 5.
Cytotoxicity and therapeutic index of selected highly bactericidal HsrA inhibitors. (A) Cytotoxicity of HsrA inhibitors I, IV, V, VIII, XI, and XII toward HeLa cells was assessed at 24 h of exposure through the PrestoBlueTM method. Experiments were performed twice in triplicate, vertical bars represent standard deviations. (B) The 50% cytotoxic concentration (CC50) was defined as the ligand concentration that reduced the viability of DMSO (vehicle) - treated cell cultures by 50%. Therapeutic index (TI) of each compound was calculated as the ratio between CC50 and MIC.
Figure 5.
Cytotoxicity and therapeutic index of selected highly bactericidal HsrA inhibitors. (A) Cytotoxicity of HsrA inhibitors I, IV, V, VIII, XI, and XII toward HeLa cells was assessed at 24 h of exposure through the PrestoBlueTM method. Experiments were performed twice in triplicate, vertical bars represent standard deviations. (B) The 50% cytotoxic concentration (CC50) was defined as the ligand concentration that reduced the viability of DMSO (vehicle) - treated cell cultures by 50%. Therapeutic index (TI) of each compound was calculated as the ratio between CC50 and MIC.
Figure 6.
Predicted model of molecular interaction between HsrA and ligand V. (A) 3D view (molecular surface) and (B) ribbon diagram of the HsrA effector domain and the best-ranked docking pose of ligand V. The helix-turn-helix (HTH) DNA binding motif has been highlighted in blue. (C) Detailed view of the amino acid residues directly involved in the interaction of HsrA with ligand V. Hydrophobic interactions are indicated with yellow dashed lines, H-bonds and halogen bonds are indicated with orange dashed lines.
Figure 6.
Predicted model of molecular interaction between HsrA and ligand V. (A) 3D view (molecular surface) and (B) ribbon diagram of the HsrA effector domain and the best-ranked docking pose of ligand V. The helix-turn-helix (HTH) DNA binding motif has been highlighted in blue. (C) Detailed view of the amino acid residues directly involved in the interaction of HsrA with ligand V. Hydrophobic interactions are indicated with yellow dashed lines, H-bonds and halogen bonds are indicated with orange dashed lines.
Figure 7.
Transcriptomic analysis of H. pylori cells exposed to lethal concentrations of HsrA bactericidal inhibitor V. Fresh cultures of H. pylori 26695 adjusted to 107 CFU/mL were exposed in triplicate to 4 × MIC of HsrA inhibitor V and incubated under controlled conditions (microaerobiosis, 37 °C) until log10 CFU/mL resulted diminished in one unit. (A) Heat map showing hierarchical cluster analysis of selected differentially expressed genes (DEG, absolute log2 fold change > 1 and p-value < 0.05). Each row represents a gene, and each column represents a sample. Red and green indicate up-regulated and down-regulated genes, respectively. The gradient colour barcode at the top right indicates the log10 normalized hit counts. (B) Volcano plot indicating distribution of DEGs. (C) Distribution of up-regulated (column in red) and down-regulated (column in blue) known-function genes by functional categories. (D) qPCR analysis of selected genes. Dashed grey lines indicate fold change cut-off values of +/- 1.5.
Figure 7.
Transcriptomic analysis of H. pylori cells exposed to lethal concentrations of HsrA bactericidal inhibitor V. Fresh cultures of H. pylori 26695 adjusted to 107 CFU/mL were exposed in triplicate to 4 × MIC of HsrA inhibitor V and incubated under controlled conditions (microaerobiosis, 37 °C) until log10 CFU/mL resulted diminished in one unit. (A) Heat map showing hierarchical cluster analysis of selected differentially expressed genes (DEG, absolute log2 fold change > 1 and p-value < 0.05). Each row represents a gene, and each column represents a sample. Red and green indicate up-regulated and down-regulated genes, respectively. The gradient colour barcode at the top right indicates the log10 normalized hit counts. (B) Volcano plot indicating distribution of DEGs. (C) Distribution of up-regulated (column in red) and down-regulated (column in blue) known-function genes by functional categories. (D) qPCR analysis of selected genes. Dashed grey lines indicate fold change cut-off values of +/- 1.5.
Table 1.
Chemical structure and physicochemical properties of relevant HsrA ligands identified by a fluorescent thermal shift-based HTS of the Maybridge HitFinderTM chemical library.
Table 2.
Minimal inhibitory and bactericidal concentrations of HsrA inhibitors against different strains of H. pylori.
Table 2.
Minimal inhibitory and bactericidal concentrations of HsrA inhibitors against different strains of H. pylori.
Inhibitor or drug |
MIC (MBC), mg/L |
H. pylori ATCC 700392 (26695)
|
H. pylori
ATCC 43504 (MTZ-R)
|
H. pylori
ATCC 700684 (CLR-R)
|
H. pylori
Donostia 2 (LVX-R)
|
I |
4 (4) |
1 (1) |
4 (4) |
2 (2) |
IV |
2 (4) |
2 (4) |
4 (4) |
4 (4) |
V |
0.063 (0.125) |
0.125 (0.125) |
0.031 (0.031) |
0.125 (0.125) |
VIII |
1 (2) |
1 (2) |
1 (2) |
2 (2) |
IX |
8 (16) |
4 (8) |
4 (8) |
8 (16) |
X |
8 (16) |
8 (8) |
8 (8) |
8 (16) |
XI |
4 (4) |
2 (4) |
2 (4) |
2 (4) |
XII |
2 (2) |
0.5 (0.5) |
0.5 (0.5) |
1 (1) |
XVII |
16 (16) |
4 (8) |
8 (8) |
8 (8) |
XXVI |
16 (16) |
8 (8) |
8 (8) |
8 (16) |
XXVIII |
32 (64) |
16 (16) |
32 (32) |
32 (32) |
XXXI |
> 64 (> 64) |
64 (64) |
> 64 (> 64) |
> 64 (> 64) |
MTZ |
1 (2) |
64 (128) |
1 (2) |
8 (8) |
CLR |
< 0.03 (< 0.03) |
< 0.03 (< 0.03) |
16 (32) |
< 0.03 (< 0.03) |
LVX |
0.12 (0.12) |
0.5 (0.5) |
0.12 (0.12) |
16 (32) |
Table 3.
Interaction of selected bactericidal HsrA inhibitors with first-line antibiotics against H. pylori ATCC 700392.
Table 3.
Interaction of selected bactericidal HsrA inhibitors with first-line antibiotics against H. pylori ATCC 700392.
Antibiotic1 |
HsrA inhibitor |
FICantibiotic |
FICinhibitor |
FICI2 |
Interaction3 |
CLR |
I |
1 |
1 |
2 |
neutral |
IV |
1 |
1 |
2 |
neutral |
V |
0.5 |
0.06 |
0.56 |
additive |
VIII |
0.5 |
0.03 |
0.53 |
additive |
XI |
0.25 |
0.5 |
0.75 |
additive |
XII |
0.25 |
0.5 |
0.75 |
additive |
MTZ |
I |
0.125 |
0.5 |
0.625 |
additive |
IV |
1 |
1 |
2 |
neutral |
V |
0.125 |
0.5 |
0.625 |
additive |
VIII |
0.5 |
0.125 |
0.625 |
additive |
XI |
0.5 |
0.063 |
0.563 |
additive |
XII |
0.25 |
0.25 |
0.5 |
synergism |
LVX |
I |
1 |
1 |
2 |
neutral |
IV |
1 |
1 |
2 |
neutral |
V |
1 |
1 |
2 |
neutral |
VIII |
1 |
1 |
2 |
neutral |
XI |
1 |
1 |
2 |
neutral |
XII |
0.5 |
0.5 |
1 |
neutral |
Table 4.
Antimicrobial activities of relevant HsrA inhibitors against C. jejuni and several representative members of normal human microbiota.
Table 4.
Antimicrobial activities of relevant HsrA inhibitors against C. jejuni and several representative members of normal human microbiota.
Inhibitor or drug |
MIC (MBC), mg/L |
C. jejuni
ATCC 33560
|
E. coli
ATCC 25922
|
K. pneumoniae ATCC 700603
|
E. faecalis
ATCC 29212
|
S. aureus
ATCC 29213
|
S. epidermidis
ATCC 12228
|
S. agalactiae
ATCC 12386
|
I |
32 (32) |
>64 (>64) |
>64 (>64) |
>64 (>64) |
>64 (>64) |
>64 (>64) |
32 (64) |
IV |
64 (>64) |
>64 (>64) |
>64 (>64) |
>64 (>64) |
>64 (>64) |
>64 (>64) |
>64 (>64) |
V |
0.25 (0.5) |
>64 (>64) |
>64 (>64) |
>64 (>64) |
>64 (>64) |
>64 (>64) |
>64 (>64) |
VIII |
16 (32) |
>64 (>64) |
>64 (>64) |
64 (64) |
16 (32) |
4 (4) |
16 (32) |
XI |
>64 (>64) |
>64 (>64) |
>64 (>64) |
64 (64) |
16 (32) |
16 (64) |
8 (8) |
XII |
64 (>64) |
>64 (>64) |
>64 (>64) |
>64 (>64) |
>64 (>64) |
>64 (>64) |
>64 (>64) |
LVX |
< 0.12 (< 0.12) |
N.D. |
0.5 (0.5) |
N.D. |
N.D. |
N.D. |
N.D. |
AMP |
N.D. |
4 (4) |
N.D. |
4 (4) |
2 (4) |
4 (4) |
0.25 (0.5) |
Table 5.
Thermodynamic parameters and interacting amino acid residues of selected HsrA-inhibitor complexes, according to ITC and molecular docking analyses.
Table 5.
Thermodynamic parameters and interacting amino acid residues of selected HsrA-inhibitor complexes, according to ITC and molecular docking analyses.
Inhibitor |
ITC1
|
Molecular Docking2
|
n |
Kd (μM) |
ΔH (kcal/mol) |
ΔG (kcal/mol) |
Interacting Residues |
I |
1.0 |
13 |
–1.2 |
–6.7 |
I121, I123, L126, I135, L152, L155, A156, R159, M195, L199, T203 |
IV |
0.9 |
3.9 |
–1.6 |
–7.4 |
I135, V142, V144, K145, G146, P148, L152, K194, P198 |
V |
1.0 |
13 |
–1.9 |
–6.7 |
L126, I135, L152, K194, M195, P198, L199, T203 |
VIII |
0.9 |
10 |
0.8 |
–6.8 |
V144, K145, G146, F149, L152, I191, K194, M195 |
XI |
1.1 |
49 |
–3.1 |
–5.9 |
I123, I128, L152, R159, D160, M195, L199, T203, C215, Y216 |
XII |
1.0 |
43 |
0.8 |
–6.0 |
I123, L126, I128, I135, L152, M195, P198, L199, T203 |