Skip to main content

Sub-lethal concentration of a colloidal nanosilver formulation (SilversolĀ®) triggers dysregulation of iron homeostasis and nitrogen metabolism in multidrug resistant Pseudomonas aeruginosa

Abstract

Background

Pseudomonas aeruginosa is a notorious pathogen. Its multidrug resistant strains are listed among priority pathogens against whom discovery of novel antibacterial agents and, elucidation of new anti-pathogenicity mechanisms are urgently warranted. This study describes multiple antibacterial effects of a colloidal nano-silver formulation- SilversolĀ® against a multi-drug resistant strain of P. aeruginosa.

Results

Minimum inhibitory concentration (MIC) of SilversolĀ® against P. aeruginosa was found to be 1.5 ppm; and at sub-MIC of 1 ppm, it was able to alter quorum-sensing regulated pigmentation (pyocanin 82%ā†“; pyoverdine 48%ā†‘), exopolysaccharide synthesis (76%ā†‘) and biofilm formation, susceptibility to antibiotics (streptomycin and augmentin), protein synthesis and export (65%ā†‘), nitrogen metabolism (37%ā†‘ nitrite accumulation), and siderophore production in this pathogen. Network analysis of the differentially expressed genes in the transcriptome of the silversol-treated bacterium identified ten genes as the potential molecular targets: norB, norD, nirS, nirF, nirM, nirQ, nosZ, nosY, narK1, and norE (all associated with nitrogen metabolism or denitrification). Three of them (norB, narK1, and norE) were also validated through RT-PCR.

Conclusions

Generation of nitrosative stress and disturbance of iron homeostasis were found to be the major mechanisms associated with anti-Pseudomonas activity of SilversolĀ®.

Peer Review reports

Introduction

Silver has a long history of therapeutic use in traditional medicine. In the modern era too, many reports [1,2,3] have been published describing various biological activities of silver. Antimicrobial activity of different forms of silver are also well known e.g., antibacterial [4, 5], antifungal [6], antiviral [7], antiprotozoal [8], and anthelmintic [9]. Different forms of silver (e.g., metallic, colloidal, silver salts) may have different modes of action, and varying degree of biological activity. Antibacterial activity of silver becomes even more important in face of the resistance displayed by bacterial pathogens against conventional antibiotics. Till now, there is not much indication from literature about bacteria developing resistance against silver. In general, silver is believed to be killing bacteria by altering cell membrane permeability, generating reactive oxygen species (ROS), and interrupting DNA replication [10,11,12,13,14]. However, much investigation still is required to elucidate precise molecular mechanisms associated with antibacterial activity of silver.

Since metallic form of silver is known to be more potent than its ionic form [15], we chose a solution of colloidal silver for the purpose of this study. Target pathogen for this study was Pseudomonas aeruginosa, one of the most notorious and versatile bacterial pathogens. Its antibiotic-resistant strains have been listed by Centers for Disease Control and Prevention (CDC; https://www.cdc.gov/drugresistance/biggest-threats.html), World Health Organization (WHO; https://www.who.int/publications/i/item/WHO-EMP-IAU-2017.12), and Department of Biotechnology of the Indian government (DBT; https://dbtindia.gov.in/sites/default/files/IPPL_final.pdf) among priority pathogens, against whom there is an urgent need to discover new antibiotics [16]. This study investigated effect of SilversolĀ® (a colloidal nano-silver formulation) on P. aeruginosaā€™s growth, various virulence traits, and gene expression profile at the whole transcriptome level. Silversol is being used primarily for multiple therapeutic applications e.g. management of oral health [17], skin diseases, burns, diabetic ulcer, and wound-care/ wound-disinfection (https://www.rxsilver.com/index_htm_files/ABLSilversafety.pdf). P. aeruginosa being one of the common wound-isolates [18, 19], we attempted to gain insight into Silversolā€™s antibacterial activity against this pathogen.

Methods

Colloidal silver formulation

The test formulation SilversolĀ® (32 ppm; Batch number: Sil Water/32P/19ā€“61) originally developed by American Biotech Labs (USA) was procured from Viridis BioPharma Pvt Ltd, Mumbai, India. It is a colloidal silver preparation reported to possess multiple biological activities [15]. The elemental form of zero-valent metallic silver particles contained in this product is coated with silver oxide, and the particle size is claimed by the manufacturer to range between 5ā€“50 nm.

Bacterial culture

The P. aeruginosa strain used in this study was sourced from our internal culture collection. This strain has been well characterized by us with respect to its antibiotic resistance, pigment production and certain other virulence traits. Its antibiogram generated through a disc-diffusion assay performed as per NCCLS guidelines revealed it to be resistant to 8 antibiotics (co-trimoxazole, augmentin, nitrofurantoin, ampicillin, chloramphenicol, clindamycin, cefixime, and vancomycin) belonging to 5 different classes (Fluoroquinolones, Beta lactams, Third generation cephalosporins, Macrolides, and Sulfonamides). Hence it can be described as a multidrug resistant (MDR) strain. As reported in our earlier publications [20, 21] involving this strain, it is a haemolytic strain capable of producing the quorum sensing (QS) regulated pigments (pyocyanin and pyoverdine), and also of biofilm formation.

As we also had P. aeruginosa PAO1 (MTCC 3541) strain in our lab, we could compare the MDR strain used in this study with the PAO1 in terms of their antibiotic resistance and virulence towards Caenorhabditis elegans. The strain used in our experiments was resistant to more classes of antibiotics, and it also displayed higher virulence towards the model host Caenorhabditis elegans (Table S1; Figure S1).

This bacterium was maintained on Pseudomonas agar (HiMedia). While culturing the bacterium for different assays, it was grown in Pseudomonas broth (magnesium chloride 1.4Ā g/L, potassium sulphate 10Ā g/L, peptic digest of animal tissue 20Ā g/L, pH 7.0ā€‰Ā±ā€‰0.2) supplemented with glycerol (3%v/v; HiMedia). Inoculum was added at 10%v/v, whose OD625 was adjusted between 0.08 and 0.10 to achieve equivalence to McFarland turbidity standard 0.5.

Quantification of growth and quorum-regulated pigments

Effect of Silversol on bacterial growth and pigment formation was quantified through broth dilution assay. Following incubation (with intermittent shaking) of P. aeruginosa in Pseudomonas broth supplemented with or without Silversol for 21ā€‰Ā±ā€‰1Ā h at 35Ā Ā°C, cell density was measured at 764Ā nm (Agilent Cary 60 UV-Vis). Pigments from the culture broth were extracted as described in [22] and [23]. One mL of culture broth was mixed in a 2:1 ratio with chloroform (Merck, Mumbai), followed by centrifugation (15,300Ā g) for 10Ā min. This resulted in formation of two immiscible layers. OD of the upper aqueous layer containing the yellow-green fluorescent pigment pyoverdine was measured at 405Ā nm. Pyoverdine Unit was calculated as OD405/OD764. The lower chloroform layer containing the blue pigment pyocyanin was mixed with 0.1Ā N HCL (20%v/v; Merck). This caused a change of colour from blue to pink. This was followed by centrifugation (15,300Ā g) for 10Ā min, and OD of upper layer acidic liquid containing pyocyanin was quantified at 520Ā nm. Pyocyanin Unit was calculated as OD520/OD764.

The lowest concentration of silver capable of inhibitingā€‰ā‰„ā€‰80% growth was taken as minimum inhibitory concentration (MIC) [24]. From each tube showing absence of growth, 0.1 mL of broth was spread on Pseudomonas agar, and these agar plates were observed for appearance of growth over an incubation (at 35ĀŗC) period of 72Ā h for determination of minimum bactericidal concentration (MBC). Extended incubation till 72Ā h was made in this case to be able to differentiate true bactericidal effect from any possible post-antibiotic effect [25, 26]. The concentration of silver which could inhibit appearance of growth on agar plates completely was taken as MBC.

Haemolysis assay

Haemolytic activity is considered to be an important virulence trait of pathogens including P. aeruginosa, particularly under iron-limiting conditions. To investigate whether silver-exposure can have any impact on haemolytic potential of P. aeruginosa, we inoculated the silver-pre-treated cells on blood agar plate (HiMedia) and compared the haemolysis pattern with that of control.

OD764 of P. aeruginosa grown in Pseudomonas broth in presence or absence of Silversol was adjusted to 1.0, and 20 ĀµL of this culture suspension was added onto the center of blood agar plate, followed by incubation at 35ĀŗC for 24Ā h. Next day plates were observed for a zone of haemolysis surrounding the point of inoculation.

Antibiotic susceptibility assay

Antibiogram of P. aeruginosaā€™s overnight grown culture in Pseudomonas broth in presence or absence of Silversol was generated through disc diffusion assay in accordance to Clinical and Laboratory Standards Institute (CLSI) guidelines [27]. Cells grown in Pseudomonas broth were separated through centrifugation (13,600Ā g) and washed with phosphate buffer (pH 7.0ā€‰Ā±ā€‰0.2) followed by centrifugation. The resulting cell pellet was used to prepare inoculum for subsequent disc diffusion assay by suspending the cells in normal saline and adjusting the OD625 between 0.08 and 0.10 to achieve an inoculum density equivalent to McFarland standard 0.5. One hundred ĀµL of this inoculum was spread onto cation-adjusted Muller-Hinton agar (HiMedia) plates (Borosil; 150Ā mm) followed by placing the antibiotic discs (Icosa G-I MINUS; HiMedia, Mumbai) on the agar surface. Incubation at 35ĀŗC was made for 18ā€‰Ā±ā€‰1Ā h, followed by observation and measurement of zone of inhibition.

Biofilm assay

Biofilm formation is an important virulence trait, and hence effect of Silversol on biofilm forming ability of P. aeruginosa, as well as on pre-formed biofilm was investigated. A flow diagram depicting all four different biofilm assays is included in supplementary file (Figure S2). Biofilm quantification was achieved through crystal violet assay [28]. Biofilm viability was assessed through MTT assay [29].

For the crystal violet assay, the biofilm containing tubes (after discarding the inside liquid) were washed with phosphate buffer saline (PBS) in order to remove all non-adherent (planktonic) bacteria, and air-dried for 15Ā min. Then, each of the washed tubes was stained with 1.5 mL of 0.4% aqueous crystal violet (Central Drug House, Delhi) solution for 30Ā min. Afterwards, each tube was washed twice with 2 mL of sterile distilled water and immediately de-stained with 1.5 mL of 95% ethanol. After 45Ā min of de-staining, 1 mL of de-staining solution was transferred into separate tubes, and read at 580Ā nm (Agilent Cary 60 UV-Vis).

For the MTT assay, the biofilm-containing tubes (after discarding the inside liquid) were washed with PBS in order to remove all non-adherent (planktonic) bacteria, and air-dried for 15Ā min. Then 1.8 mL of minimal media (sucrose 15Ā g/L, K2HPO4 5Ā g/L, NH4Cl 2Ā g/L, NaCl 1Ā g/L, MgSO4 0.1Ā g/L, yeast extract 0.1Ā g/L, pH 7.4ā€‰Ā±ā€‰0.2) was added into each tube, followed by addition of 200 ĀµL of 0.3% MTT [3-(4,5- Dimethylthiazol-2-yl)-2,5- iphenyltetrazolium Bromide; HiMedia]. Then after 2Ā h incubation at 35Ā Ā°C, all liquid content was discarded, and the remaining purple formazan derivatives were dissolved in 2 mL of DMSO and measured at 540Ā nm.

Exopolysaccharide (EPS) quantification

P. aeruginosa was grown in 100 mL flasks containing 20 mL of Pseudomonas broth. Incubation at 35ĀŗC was made for 24Ā h with intermittent shaking. Following estimation of growth by measuring OD at 764Ā nm, culture broth was subjected to centrifugation (13,600Ā g for 10Ā min), and the supernatant was used for EPS quantification using the method described in [30] with some modification. Briefly, 40 mL of chilled acetone (Merck) was added to 20 mL of supernatant, and allowed to stand for 30Ā min. The EPS precipitated thus was separated by filtration through pre-weighed Whatman # 1 filter paper (Whatman International Ltd., England). Filter paper was dried at 60Ā° C for 24Ā h, and weight of EPS on paper was calculated.

Protein estimation

Extracellular protein present in bacterial culture (grown in presence or absence of silver for 21ā€‰Ā±ā€‰1Ā h at 35Ā Ā°C) supernatant, and intracellular protein in the cell lysate was quantified through Folin-Lowry method [31, 32]. After measuring cell density, 1 mL of P. aeruginosa culture grown in Pseudomonas broth was centrifuged (13,600Ā g), and the resulting supernatant was used for extracellular protein estimation. The remaining cell pellet was subjected to lysis [33] for release of intracellular proteins. Briefly, the cell pellet was washed with phosphate buffer (pH 7.4), and centrifuged (13,600Ā g). Resulting pellet was resuspended in 1 mL of chilled lysis buffer (8.76Ā g/L NaCl, 10 mL of Triton X 100, 5/L g sodium deoxycholate, 1Ā g/L sodium dodecyl sulphate, and 6Ā g/L Tris HCl, in 990 mL of distilled water), and centrifuged (500Ā rpm) for 30Ā min at 4Ā Ā°C for agitation purpose. This was followed by further centrifugation (16,000Ā g at 4Ā Ā°C) for 20Ā min. Resulting cell lysate (supernatant) was used for protein estimation. Kanamycin (at IC50: 200Ā Āµg/mL), an aminoglycoside antibiotic known to inhibit bacterial protein synthesis [34], was used as a positive control.

Nitrite estimation

Quantification of nitrite in bacterial culture was achieved through a colorimetric assay using modified Griess reagent [35]. A total of 250 ĀµL of supernatant obtained from centrifugation (13,500Ā g for 10Ā min at 25Ā Ā°C) of P. aeruginosa culture grown in presence or absence of silver (for 21ā€‰Ā±ā€‰1Ā h at 35Ā Ā°C), was mixed with an equal volume of Griess reagent (1X concentration; Sigma-Aldrich) followed by 15Ā min incubation in dark at room temperature. Absorbance of the resulting colour was measured at 540Ā nm. This absorbance was plotted on the standard curve prepared using NaNO2 (0.43-65 ĀµM) to calculate nitrite concentration. Sodium nitroprusside (Sigma Aldrich) was used as a positive control, as it is known to generate nitrosative stress in bacteria [36].

Transcriptome analysis

To gain insights into the molecular mechanisms through which silver could inhibit bacterial growth and modulate various traits like QS, gene expression profile of P. aeruginosa challenged with sub-MIC of Silversol (1 ppm) was compared with that of control culture at the whole transcriptome level. Overall workflow of this whole transcriptome analysis (WTA) aimed at obtaining a holistic picture regarding mode of action of this formulation is presented in Figure S3.

RNA extraction and library preparation

RNA from bacterial cells was extracted by Trizol (Invitrogen Bioservices; 343,909) method, and dissolved in nuclease free water, followed by purity and concentration check, and quantifying RIN (RNA Integrity Number) values. Final libraries were quantified through Qubit 4.0 fluorometer (Thermofisher; Q33238). RIN values and acquired sizes of all libraries are reported in Table S2.

Genome annotation and functional analysis

Quality assessment of the raw fastq reads of the sample was performed using FastQC v.0.11.9 (default parameters) [37] and Fastp v.0.20.1 [38]. The P. aeruginosa genome (GCA_000006765.1_ASM676v1) was indexed using bowtie2-build [39] v2.4.2 (default parameters), and the processed reads were mapped to it using bowtie2 v2.4.2 parameters. The aligned reads from the individual samples were quantified using feature count v. 0.46. 1 [40] to obtain gene counts. These gene counts were used for differential expression estimation. The up- and down-regulated sequences were extracted from the P. aeruginosa coding file and subjected to blast2go [41] for annotation to extract the Gene Ontology (GO) terms. These GO terms were subjected to the wego [42] tool to obtain gene ontology bar plots.

All the raw sequence data has been submitted to Sequence Read Archive. Relevant accessions no. is SRX14392191 (https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/sra/SRX14392191).

Network analysis

From the list of differentially expressed genes (DEG) in Silversol-exposed P. aeruginosa, those satisfying dual filter criteria of log fold-changeā€‰ā‰„ā€‰2 and FDRā€‰ā‰¤ā€‰0.05 were chosen for further network analysis. The list of such DEGs was fed into the STRING (v.11.5) database [43] to generate the PPI (Protein-Protein Interaction) network. Members of this PPI network were then arranged in decreasing order of ā€˜node degreeā€™ (a measure of connectivity with other genes or proteins), and those above a specified threshold value were subjected to ranking by the cytoHubba plugin (v.3.9.1) [44] of Cytoscape [45]. As cytoHubba employs 12 different ranking methods, we considered the DEG being top-ranked by ā‰„ā€‰6 different methods (i.e., 50% of the total ranking methods) for further investigation. These top-ranked shortlisted proteins were then subjected to local cluster analysis through STRING, and those that were part of multiple clusters were termed potential ā€˜hubsā€™ that can be investigated for additional validation of their targetability. The term ā€˜hubā€™ refers to a gene or protein that interacts with multiple other genes/proteins. The identified hubs were then subjected to co-occurrence analysis to see whether an antibacterial agent targeting them is likely to meet the requirement of selective toxicity (targeting the pathogen while causing no harm to the host). This sequence of analysis allowed us to end up with a limited number of proteins that satisfied multiple statistical and biological significance criteria simultaneously: (i) log fold-changeā€‰ā‰„ā€‰2; (ii) FDRā€‰ā‰¤ā€‰0.05; (iii) relatively higher node degree; (iv) top-ranking by at least 6 cytoHubba methods; (v) (preferably) member of more than 1 local network cluster; and (vi) high probability of the target being absent from the host.

Polymerase chain reaction (RT-PCR)

Differential expression of the potential hubs identified through network analysis of DEG revealed from WTA was confirmed through PCR too. Primer designing for the selected genes was accomplished through Primer3 Plus [46]. Primer sequences thus obtained were confirmed for their binding withing the whole P. aeruginosa genome exclusively to the target gene sequence. Primer sequences for all the target genes are listed in TableĀ 1. RNA extraction and quality check was done as described in preceding section. cDNA was obtained by using SuperScriptā„¢ VILOā„¢ cDNA Synthesis Kit (Invitrogen Biosciences). PCR assay was conducted by using gene specific primers procured from Sigma-Aldrich. The gene PA3617 (recA) was kept as an endogenous control. The reaction mix used was FastStart Essential DNA Green Master mix (Roche; 06402712001). Real time PCR assay was performed on Quant studio 5 real time PCR machine (Thermo Fisher Scientific, USA). Temperature profile employed is provided in Table S3.

Table 1 Primer sequences for the target genes

Statistics

All results reported are means of three or more independent experiments, each performed in triplicate. Statistical significance was assessed through t-test performed using Microsoft ExcelĀ®, and data with pā€‰ā‰¤ā€‰0.05 was considered to be significant.

Results and discussion

Silver inhibits growth of P. aeruginosa and modulates QS-regulated pigment formation

P. aeruginosa was challenged with different Silversol concentrations (0.03- 2 ppm). Bacterial growth and pyoverdine production remained unaffected till 0.5 ppm Silversol, while pyocyanin formation was inhibited in a dose-dependent fashion 0.5 ppm onward (Fig.Ā 1A). Effect of Silversol on pyoverdine production appeared to follow an inverted U-shaped hormetic dose response curve [47] between the concentration range 0.5ā€“1.5 ppm. Since Silversol inhibited pyocyanin formation at 0.5 ppm without affecting bacterial growth, this concentration can be said to be purely quorum modulatory. Since 1 ppm seemed to be the ~ā€‰IC50, and this concentration of Silversol also had a heavy effect on both quorum-regulated pigments, we chose this as the test concentration for further experiments. When growth curve of P. aeruginosa grown in presence of this sub-MIC concentration of Silversol (1 ppm) was compared with that grown in absence of Silversol over a period of 48-hour (Fig.Ā 1B), it was observed that Silversol causes a delay in appearance of visible growth, and it allows cell density to reach only half of that of control culture till 27Ā h, however after that the difference between cell density of control and experimental culture starts getting narrow, and after 48Ā h of incubation, the cell density in both cultures gets at par statistically. This pattern of delayed onset of observed parameter in presence of Silversol, followed by recovery at par to the control culture in later phase of growth curve was observed in case of pigment production too [Figure S5 (A-B)], which is not surprising as production of both the pigments quantified here is regulated in a cell density-dependent manner in P. aeruginosa [48].

While Silversol seemed to inhibit pigment production completely 1.5 ppm onward, and caused complete visible inhibition of growth 2 ppm onward, the MBC was found to be somewhere between 16 and 20 ppm (Figure S4). When cells from experimental tubes were plated onto Pseudomonas agar (not containing any Silversol), only those coming from 1 ppm silver tubes gave pigmented growth, while those from 10ā€“15 ppm silver tubes grew without pigmentation. Inoculum coming from the 20 ppm silver tubes failed to give rise to any growth on agar plates indicating that MBC has been reached.

Silver appears to disturb iron homeostasis of P. aeruginosa

Silversol (1 ppm) enhanced pyoverdine production by almost 1.5-fold and inhibited pyocyanin heavily. Since pyoverdine is a siderophore [49], and pyocyanin also plays an important role in iron-sequestration [50, 51], we speculated that Silversol might be disturbing iron-homeostasis in P. aeruginosa. To have additional insight into this aspect, we inoculated blood agar plates with silver-pre-exposed P. aeruginosa, and these cells were found to be more haemolytic than their silver-not-exposed counterparts (Fig.Ā 1C). Taking the effect of silver on pyoverdine production and haemolytic activity of the pathogen together, it can be said that silver-exposed cells seem to experience iron-limitation and to overcome this the cells are producing more siderophore and haemolysins. To further confirm the iron homeostasis-disturbing activity of Silversol, we grew P. aeruginosa in Pseudomonas broth supplemented with Silversol as well as different concentrations of ferrous sulphate (Merck) to check whether sufficient iron supply can protect P. aeruginosa from Silversolā€™s antibacterial effect. Indeed, P. aeruginosa was able to remain resistant to Silversolā€™s growth-inhibitory effect in presence of FeSO4, and it also produced lesser pyoverdine than when growing in Pseudomonas broth with or without Silversol [Figure S5(C)]. It can be said that in presence of additional iron in growth media, the bacterium is not forced to make extra efforts for iron scavenging in terms of pyoverdine production, and hence Silversolā€™s negative effect on bacterial iron-homeostasis is confirmed, and this effect may be much more detrimental in iron-limiting conditions inside a host. Most microbes need iron at a concentration of 10āˆ’ā€‰6Ā M for their growth and reproduction, while only minute quantities (~ā€‰10ā€“15Ā M) of free iron are available in the human body [52].

Silver-treated P. aeruginosa displays higher susceptibility to augmentin and streptomycin

To investigate whether silver exposure influences antibiotic susceptibility of P. aeruginosa, we grew P. aeruginosa in presence of 1 ppm Silversol, and the antibiogram of the resulting cells (after washing with phosphate buffer) was compared to that of control cells. Silver-pre-exposed cells displayed a phenotype change from ā€˜resistantā€™ to ā€˜sensitiveā€™ against augmentin; and a notable increase (38%) in susceptibility to streptomycin (TableĀ 2; Fig.Ā 1D). Such resistance-modifying activity of the silver becomes quite important given P. aeruginosa is known rarely to be susceptible to augmentin [53], and augmentin resistance is quite prevalent among its clinical isolates [54]. Streptomycin-resistance was reported by [55] to be among the most common resistance patterns in P. aeruginosa clinical isolated from Mexico.

Silversol could partially eradicate pre-formed P. aeruginosa biofilm

While Silversol inhibited biofilm formation in P. aeruginosa partially (19%), it could also eradicate the pre-formed biofilm (by 34%) and reduce biofilm viability by 30%. Inhibitory effect of Silversol on P. aeruginosa biofilms seemed independent from its growth-inhibitory effect, since Silversol-pre-treated cells subsequently allowed to form biofilm in test-tubes filled with Silversol-free growth media, could form 43% lesser biofilm (Fig.Ā 2A). Since bacterial biofilms are known for displaying higher antibiotic resistance than their planktonic counterparts [56], biofilm-eradicating agents can help restoring the antibiotic susceptibility of bacterial cells by freeing them of the biofilm matrix.

Since exopolysaccharides (EPS) constitute an important component of bacterial biofilm matrix, we quantified EPS in control vs. silver-treated P. aeruginosa culture. Though the absolute EPS in experimental and control cultures was at par, nullifying it against reduced cell density in silver-exposed culture, EPS production in experimental culture was found to be enhanced by 76.34% (Fig.Ā 2B). Higher EPS synthesis can be considered as an indication of envelope stress [57] in the silver-exposed bacteria. When faced with a variety of stress conditions, P. aeruginosa is known to respond by escalating its EPS synthesis as a survival strategy, since EPS serves as a protective matrix [58, 59], whose production is upregulated during nutrient limitation and oxidative stress [60, 61]. This study found the sub-MIC level of Silversol to disturb QS-regulated traits in P. aeruginosa such as pigmentation [Fig.Ā 1A and Figure S5(A)-(B)] and biofilm formation (Fig.Ā 2A), and the quorum sensing system in P. aeruginosa is known to influence EPS production too [62]. Studying the correlation between EPS production and stress-response in P. aeruginosa can provide new insights into its survival mechanisms and pathogenicity.

P. aeruginosa seemed to upregulate its protein synthesis in presence of silver

P. aeruginosa exposed to sub-MIC of Silversol was found to have higher intracellular as well as extracellular protein concentration than its silver-non-exposed counterpart (Fig.Ā 2C). Bacterial response to the known inhibitor of protein synthesis (Kanamycin) was also similar. It may be said that bacteria responds to the inhibitory effect of such antimicrobials exerting their action through suppressing protein synthesis, by upregulating its protein sysnthesis and/or secretion machinery to compensate the inhibitory effect of the antimicrobial agents [63]. Such upregulation of protein synthesis might have stemmed from the translational reprogramming in the stressed cells [64], as they are facing the stress of the antibacterial activity of the silver.

Fig. 1
figure 1

(A) Growth-inhibitory and quorum-modulatory effect of Silversol on P. aeruginosa. Bacterial growth was measured as OD764; OD of pyoverdine and pyocyanin was measured at 405Ā nm and at 520Ā nm respectively. Pyoverdine Unit and Pyocyanin Unit were calculated as the ratio OD405/OD764 and OD520/OD764(an indication of pigment production per unit of growth); Ofloxacin (0.3Ā Āµg/mL) inhibited growth by 76**%Ā±0.1 with affecting pigment production. **pā€‰<ā€‰0.01, ***pā€‰<ā€‰0.001; minus sign (-) in parentheses indicate a decrease over control; (B) P. aeruginosa grows at slower growth rate in presence of sub-MIC of Silversol. Generation time of P. aeruginosa in presence and absence of Silversol was calculated to be 12.15Ā h and 11.17Ā h. For each time point shown in this growth curve, pigment quantification was also done. To avoid overcrowding of the graph, data pertaining to pigments is presented separately in Figure S5; (C) Silversol pre-treated P. aeruginosa exhibits enhanced haemolysis on blood agar plates. Silver-pretreated P. aeruginosa when inoculated onto blood agar plates, produced a clearer zone of haemolysis than its control counterpart receiving no previous exposure to silver; (D) Silversol pre-treatment enhances P. aeruginosaā€™s susceptibility to augmentin and streptomycin. Silver-pretreated cells seem to be altered in their response to antibiotics with respect to growth as well as QS-regulated pigmentation. On these plates of cation-adjusted Muller-Hinton agar, clear zones of inhibition can be seen surrounding the discs of augmentin and streptomycin in the experimental plate inoculated with silver-pre-treated cells, while such zones are absent (augmentin) or faint (streptomycin) from the control plate inoculated with P. aeruginosa having no previous silver-exposure

Table 2 Silver pre-treatment modulates bacterial susceptibility to some antibiotics

Silver induces nitrosative stress in P. aeruginosa

Since denitrification pathway is an important metabolic pathway in P. aeruginosa, and enzymes involved in detoxification of reactive nitrogen species are proposed to be potential anti-pathogenic targets [65, 66], we quantified one of the intermediates of denitrification pathway, nitrite (NO2-), in silver-exposed P. aeruginosa. Culture supernatant of the latter was found to possess 37% higher nitrite (Fig.Ā 2D) than that grown in absence of silver. Higher nitrite concentrations can have multiple effects on P. aeruginosa physiology and virulence. It can exert its toxicity by disrupting electron transport chain, and can also impair bacterial virulence, besides modulating susceptibility to various antibiotics. Nitrite can react with other reactive nitrogen species (RNS) to form highly reactive intermediates, such as peroxynitrite (ONOO-), which can damage cellular components, including proteins, lipids, and DNA [67]. Nitrite can interfere with the electron transport chain in P. aeruginosa, leading to a decrease in ATP production and compromised energy metabolism [68, 69]. This disruption can negatively impact various cellular processes and growth. Nitrite can also inhibit the production of virulence factors such as pyocyanin, elastase, and siderophores [70]. Nitrite build-up can negtively impact bacterial physiology by trigerring the overexpression of efflux pumps and altering membrane permeability [71].

Differential gene expression in silver-treated P. aeruginosa

Silversol-exposed P. aeruginosa when subjected to whole transcriptome analysis, it was found to express a total of 26 genes (0.48% of total genome) differently (log fold-changeā€‰ā‰„ā€‰2 and FDRā€‰ā‰¤ā€‰0.05). List of differentially expressed genes (DEG) is provided in TableĀ 3, and corresponding heat map (Figure S6) and volcano plot (Figure S7) can be seen in supplementary file. When we looked for functions of the DEG in appropriate databases like KEGG, PDB, or UniProt, all the 6 downregulated genes turned out to be hypothetical proteins. Amongst the upregulated genes in silver-exposed P. aeruginosa culture, 13 genes were associated with nitrogen metabolism, 2 were associated with heme biosynthesis or acquisition, three were hypothetical proteins, 1 was pyoverdine regulator and 1 was a protein folding catalyst. Function-wise categorisation of all DEG is presented in Figure S8.

Fig. 2
figure 2

Silver modulates biofilm formation, exopolysaccharide synthesis, protein synthesis, and nitrogen metabolism in P. aeruginosa. (A) Silversol could partially inhibit P. aeruginosa biofilm formation, and eradicate the pre-formed biofilm. Crystal violet assay was performed to quantify biofilm formation, and eradication. Cell viability in the biofilm was estimated through MTT assay; (B) Silversol enhances EPS synthesis in P. aeruginosa. Though there are lesser number of cells in the silver-supplemented media, they synthesized EPS in amount equal to their silver-unexposed counterparts. EPS Unit was calculated as Cell Density (OD764): EPS (g/L) ratio; (C)Ā P. aeruginosa grown in presence of Silversol registered higher protein synthesis. Intracellular and extracellular protein concentrations in P. aeruginosa grown in presence of Silversol at sub-MIC level were significantly higher as compared to its silver-non-exposed counterpart. Kanamycin employed as a positive control at its sub-MIC level also generated similar response from bacterial culture. Protein Unit (i.e., Protein concentration: Cell density ratio) was calculated to nullify any effect of cell density on protein production; (D) Silversol-treated P. aeruginosa culture has higher extracellular accumulation of nitrite. Silversol caused nitrite concentration in P. aeruginosa culture supernatant to rise when compared to control. Sodium nitroprusside (SNP) used as a positive control also caused higher nitrite build up in P. aeruginosa culture. Nitrite Unit (i.e., Nitrite concentration: Cell density ratio) was calculated to nullify any effect of cell density on nitrite production. *pā€‰<ā€‰0.05, **pā€‰<ā€‰0.01, ***pā€‰<ā€‰0.001, ns: non-significant

Empirically the transcriptomic profile indicated Silversol to disturb iron homeostasis and nitrogen metabolism in P. aeruginosa. The hypothesis of disturbance of iron homeostasis corroborates well with results of our in vitro experiments for assessing haemolytic potential (Fig.Ā 1C) and pyoverdine [Fig.Ā 1A and Figure S5 (C)] production. It seems that in presence of Silversol, the bacteria are facing iron-limitation and is forced to upregulate the genes coding for heme synthesis and siderophore production to maintain iron homeostasis. This observation is important in light of the fact that iron is an essential micronutrient for bacteria, and since the concentration of free iron in human serum is quite low [72], pathogens have to develop iron-scavenging strategies for a successful in-host survival. Differential expression of multiple genes associated with nitrogen metabolism also corroborates well with increased nitrite concentration in silver-treated bacterial culture supernatant (Fig.Ā 1D).

To have a holistic idea of the antibacterial mechanism of Silversol, we subjected all the DEG to network analysis through STRING. The resulting protein-protein interaction (PPI) (Fig.Ā 3A) network had 26 nodes connected through 74 edges with an average node degree of 5.69. Since the number of edges (74) in this PPI network is almost 25-fold higher than expected (03) with a PPI enrichment pā€“valueā€‰<ā€‰1.0e-16, this network can be said to possess significantly more interactions among the member proteins than what can be expected for a random set of proteins having identical sample size and degree distribution. Such an enrichment is suggestive of the member proteins being at least partially biologically connected.

Table 3 List of DEGs in Silversol exposed P. aeruginosa satisfying the dual criteria of log fold-changeā€‰ā‰„ā€‰2 and FDRā€‰ā‰¤ā€‰0.05
Fig. 3
figure 3

(A) PPI network of DEGs in Silversol-exposed P. aeruginosa;(B) PPI network of top-ranked genes revealed through cytoHubba among DEG in Silversol-exposed P. aeruginosa

When we arranged all the 26 nodes in decreasing order of node degree, 19 nodes were found to have a non-zero score, and we selected top 16 genes with a node degreeā€‰ā‰„ā€‰4 for further ranking by different cytoHubba methods. Then we looked for genes which appeared among top ranked candidates by ā‰„ā€‰6 cytoHubba methods. This allowed us to shortlist ten genes (TableĀ 4) which were ranked among top-10 by ā‰„ā€‰9 cytoHubba methods to be taken for further cluster analysis. Interaction map of these 10 important genes (Fig.Ā 3B) showed them to be networked with the average node degree score of 8. Number of edges possessed by this network was 40 as against expected 0 for any such random set of proteins. These 10 genes were found to be distributed among five different local network clusters. Strength score for each of these clusters was >ā€‰1.86. While five of the proteins (norB, PA0525, nirS, nirQ, and nosZ) were members of all the five clusters, two proteins (nosY and PA0521) were part of four different clusters. Of the remaining three, nirF was part of three different clusters, while nirM and narK1 were respectively members of two and a single cluster. The multi-cluster proteins identified here can be said as not only the most important targets of Silversol, but also hub proteins with high network centrality and potential targets for novel anti-Pseudomonas drug discovery programmes.

We also conducted a gene cooccurrence pattern analysis of gene families across genomes (through STRING) with respect to the potential hubs identified by us (Fig.Ā 4A). None of the 10 potential targets was shown to be present in humans, hence any anti-pathogenic drugs capable of causing dysregulation of these genes are least likely to toxic to humans. This corroborates well with reports showing Silversol to be safe for human consumption [15]. Three of the Silversolā€™s target genes in P. aeruginosa were also shown to be present in Staphylococcus aureus. However, except narK1, no other target proteins were shown to be present in other important pathogens. Since Silversol is already known to be active against a wide spectrum of gram-positive and gram-negative bacteria [15], it may be believed to be acting against different organisms through different mechanisms.

One of the side-effect with conventional antibiotics is that they fail to differentiate between the ā€˜goodā€™ (symbionts in human microbiome) and ā€˜badā€™ (pathogens) bacteria, and hence their therapeutic use may lead to gut dysbiosis. An ideal antimicrobial agent is expected to target pathogens exclusively without causing gut dysbiosis. In this respect, a target in pathogenic bacteria absent from symbionts of human microbiome will be most suitable candidate for antibiotic discovery programmes. To have some insight on this front with respect to the targets identified by us, we run a gene cooccurrence analysis with two representative ā€˜goodā€™ bacteria too, reported to be part of healthy human microbiome, and the said targets were not shown to be present in them. This corroborates with the selective inaction of Silversol on probiotic strains of Lactobacillus acidophilus and Bifidobacterium longum documented in internal reports of Viridis Biopharma (Selective inaction of ASAP on probiotics. Unpublished raw data, 2004).

Target validation through RT-PCR

Based on the network analysis of transcriptome of silver-treated P. aeruginosa, we selected following three genes for further validation through RT-PCR: nor B (identified as member of all the 5 clusters in network analysis), PA0521 (identified as member of 4 different clusters in network analysis), and narK1 (shown by gene cooccurrence analysis to be present across multiple pathogenic genera); wherein they were found to be up-regulated in silver-treated P. aeruginosa by 2.65-fold, 2.32-fold, and 7.37-fold respectively (Fig.Ā 4B).

Table 4 Top ten cytoHubba-ranked genes from among the top-16 with respect to node degree
Fig. 4
figure 4

(A) Cooccurrence analysis of genes coding for potential targets in P. aeruginosa; (B) Confirmation of differential expression of selected genes in Silversol-treated P. aeruginosa through RT-PCR. ***pā€‰<ā€‰0.001; ** pā€‰<ā€‰0.01

Conclusion

Silver has long been known for its antimicrobial activity. Though literature contains reports describing a variety of modes of antibacterial action of silver and its nanoparticles, our knowledge on this front is far from complete. Antibacterial mechanisms of silver previously described by various researchers include generation of oxidative stress [73,74,75], generating nitrosative stress [75], disruption of membrane integrity [76], protein denaturation [77, 78], inhibition of enzymatic activity [79, 80], interference with quorum sensing [81, 82], inducing DNA damage [74] and interfering with its replication and transcription [83], inhibition of efflux pumps [81], etc. The present study investigated effect of sub-MIC level of colloidal silver on P. aeruginosaā€™s growth, QS-regulated pigmentation, biofilm, protein synthesis, nitrogen metabolism, EPS synthesis, haemolytic activity and siderophore production, antibiotic susceptibility, and gene expression at the whole transcriptome level. A schematic summary of Silversolā€™s multiple effects on P. aeruginosa is presented in Fig.Ā 5. Disruption of iron homeostasis and generation of nitrosative stress seemed to be the major mechanisms of anti-P. aeruginosa activity of silver in this study. Differential expression of three important genes involved in denitrification pathway in silver-exposed P. aeruginosa was confirmed through RT-PCR too. Hub proteins identified in this study as major targets of silver in P. aeruginosa warrants further investigation with respect to validating their targetability e.g., by confirming defective growth of mutant strains of P. aeruginosa bearing deletion of one or more of the identified hub genes. Such validated targets can prove vital to various antibiotic discovery efforts globally. Though the gene expression profile of the bacterium under influence of sub-lethal concentrations of silver may vary from that under influence of its lethal concentrations, this study provides useful insights into antibacterial mechanism of silver and identification of its potential targets in P. aeruginosa.

Fig. 5
figure 5

Overall schematic of multiple effects of Silversol on Pseudomonas aeruginosa. Various physiological and virulence traits of P. aeruginosa affected under the influence of sub-lethal concentration of Silversol are depicted. Up (ā†‘) or down (ā†“) regulation of the genes relevant to those traits is also indicated. EPS: exopolysaccharide

Data Availability

All the data has been provided within main manuscript or supplementary files.

All the raw sequence data has been submitted to Sequence Read Archive. Relevant accessions no. is SRX14392191 (https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/sra/SRX14392191).

References

  1. Zhang XF, Liu ZG, Shen W, Gurunathan S. Silver nanoparticles: synthesis, characterization, properties, applications, and therapeutic approaches. Int J Mol Sci. 2016;17(9):1534. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms17091534.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  2. Anandaradje A, Meyappan V, Kumar I, Sakthivel N. Microbial synthesis of silver nanoparticles and their biological potential. Nanopart Med. 2020;99ā€“133. https://0-doi-org.brum.beds.ac.uk/10.1007/978-981-13-8954-2_4.

  3. Wahab MA, Luming L, Matin MA, Karim MR, Aijaz MO, Alharbi HF, Abdala A, Haque R. Silver micro-nanoparticle-based nanoarchitectures: synthesis routes, biomedical applications, and mechanisms of action. Polymers. 2021;13(17):2870. https://0-doi-org.brum.beds.ac.uk/10.3390/polym13172870.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  4. Tang S, Zheng J. Antibacterial activity of silver nanoparticles: structural effects. Adv Healthc Mater. 2018;7(13):1701503. https://0-doi-org.brum.beds.ac.uk/10.1002/adhm.201701503.

    ArticleĀ  CASĀ  Google ScholarĀ 

  5. Balachandar R, Navaneethan R, Biruntha M, Kumar KK, Govarthanan M, Karmegam N. Antibacterial activity of silver nanoparticles phytosynthesized from Glochidion candolleanum leaves. Mater Lett. 2022;311:131572. https://0-doi-org.brum.beds.ac.uk/10.1016/j.matlet.2021.131572.

    ArticleĀ  CASĀ  Google ScholarĀ 

  6. Matras E, Gorczyca A, Przemieniecki SW, Oćwieja M. Surface properties-dependent antifungal activity of silver nanoparticles. Sci Rep. 2022;12(1):18046. https://0-doi-org.brum.beds.ac.uk/10.1038/s41598-022-22659-2.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  7. Salleh A, Naomi R, Utami ND, Mohammad AW, Mahmoudi E, Mustafa N, Fauzi MB. The potential of silver nanoparticles for antiviral and antibacterial applications: a mechanism of action. Nanomaterials. 2020;10(8):1566. https://0-doi-org.brum.beds.ac.uk/10.3390/nano10081566.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  8. Hassan D, Farghali M, Eldeek H, Gaber M, Elossily N, Ismail T. Antiprotozoal activity of silver nanoparticles against Cryptosporidium parvum oocysts: new insights on their feasibility as a water disinfectant. J Microbiol Methods. 2019;165:105698. https://0-doi-org.brum.beds.ac.uk/10.1016/j.mimet.2019.105698.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  9. Gajera G, Godse C, DeSouza A, Mehta D, Kothari V. Potent anthelmintic activity of a colloidal nano-silver formulation (SilversolĀ®) against the model worm Caenorhabditis Elegans. 2023. https://0-doi-org.brum.beds.ac.uk/10.21203/rs.3.rs-2091038/v1.

  10. Ahmad A, Wei Y, Syed F, Tahir K, Rehman AU, Khan A, Ullah S, Yuan Q. The effects of bacteria-nanoparticles interface on the antibacterial activity of green synthesized silver nanoparticles. Microb Pathog. 2017;102:133ā€“42. https://0-doi-org.brum.beds.ac.uk/10.1016/j.micpath.2016.11.030.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  11. Mazur P, Skiba-Kurek I, Mrowiec P, Karczewska E, Drożdż R. Synergistic ROS-associated antimicrobial activity of silver nanoparticles and gentamicin against staphylococcus epidermidis. Int J Nanomed. 2020;3551ā€“62. https://0-doi-org.brum.beds.ac.uk/10.2147/IJN.S246484.

  12. Reeves CM, Magallon J, Rocha K, Tran T, Phan K, Vu P, Yi Y, Oakley-Havens CL, Cedano J, Jimenez V, Ramirez MS. Aminoglycoside 6ā€²-N-acetyltransferase type ib [AAC (6ā€²)-Ib]-Mediated Aminoglycoside Resistance: phenotypic Conversion to susceptibility by Silver Ions. Antibiotics. 2020;10(1):29. https://0-doi-org.brum.beds.ac.uk/10.3390/antibiotics10010029.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  13. Yin IX, Zhang J, Zhao IS, Mei ML, Li Q, Chu CH. The antibacterial mechanism of silver nanoparticles and its application in dentistry. Int J Nanomed. 2020;17:2555ā€“62. https://0-doi-org.brum.beds.ac.uk/10.2147/IJN.S246764.

    ArticleĀ  Google ScholarĀ 

  14. Jiang S, Wang F, Cao X, Slater B, Wang R, Sun H, Wang H, Shen X, Yao Z. Novel application of ion exchange membranes for preparing effective silver and copper based antibacterial membranes. Chemosphere. 2022;287:132131. https://0-doi-org.brum.beds.ac.uk/10.1016/j.chemosphere.2021.132131.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  15. de Souza A, Vora AH, Mehta AD, Moeller K, Moeller C, Willoughby AJ, Godse CS. SilverSolĀ® a Nano-Silver Preparation: A Multidimensional Approach to Advanced Wound Healing. Wound Healing Research: Current Trends and Future Directions. 2021:355 ā€“ 96. https://0-doi-org.brum.beds.ac.uk/10.1007/978-981-16-2677-7_12.

  16. Weiner LM, Webb AK, Limbago B, Dudeck MA, Patel J, Kallen AJ, Edwards JR, Sievert DM. Antimicrobial-resistant pathogens associated with healthcare-associated Infections: summary of data reported to the National Healthcare Safety Network at the Centers for Disease Control and Prevention, 2011ā€“2014. Infect Control Hosp Epidemiol. 2016;37(11):1288ā€“301.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  17. Tran PL, Luth K, Wang J, Ray C, de Souza A, Mehta D, Moeller KW, Moeller CD, Reid TW. Efficacy of a silver colloidal gel against selected oral bacteria in vitro. F1000Research. 2019;8. https://0-doi-org.brum.beds.ac.uk/10.12688/f1000research.17707.1.

  18. Reyes J, Komarow L, Chen L, Ge L, Hanson BM, Cober E, Herc E, Alenazi T, Kaye KS, Garcia-Diaz J, Li L. Global epidemiology and clinical outcomes of carbapenem-resistant Pseudomonas aeruginosa and associated carbapenemases (POP): a prospective cohort study. The Lancet Microbe. 2023;4(3):e159ā€“70. https://0-doi-org.brum.beds.ac.uk/10.1016/S2666-5247(22)00329-9.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  19. Phan S, Feng CH, Huang R, Lee ZX, Moua Y, Phung OJ, Lenhard JR. Relative abundance and detection of Pseudomonas aeruginosa from Chronic Wound Infections globally. Microorganisms. 2023;11(5):1210. https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms11051210.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  20. Joshi C, Patel P, Kothari V. Anti-infective potential of hydroalcoholic extract of Punica granatum peel against gram-negative bacterial pathogens. F1000Research. 2019;8(70):70. https://0-doi-org.brum.beds.ac.uk/10.12688/f1000research.17430.2.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  21. Patel P, Joshi C, Kothari V. Antipathogenic potential of a polyherbal wound-care formulation (herboheal) against certain wound-infective gram-negative bacteria. Adv. Pharmacol. Sci. 2019;2019. https://0-doi-org.brum.beds.ac.uk/10.1155/2019/1739868.

  22. Unni KN, Priji P, Geoffroy VA, Doble M, Benjamin S. Pseudomonas aeruginosa BUP2ā€”A novel strain isolated from malabari goat produces type 2 pyoverdine. Adv Biosci Biotechnol. 2014;5(11):874. https://0-doi-org.brum.beds.ac.uk/10.4236/abb.2014.511102.

    ArticleĀ  Google ScholarĀ 

  23. El-Fouly MZ, Sharaf AM, Shahin AA, El-Bialy HA, Omara AM. Biosynthesis of pyocyanin pigment by Pseudomonas aeruginosa. J Radiat Res Appl Sci. 2015;8(1):36ā€“48. https://0-doi-org.brum.beds.ac.uk/10.1016/j.jrras.2014.10.007.

    ArticleĀ  CASĀ  Google ScholarĀ 

  24. Turnidge JD. Susceptibility test methods: general considerations. Manual of clinical microbiology. 2015:1246-52.

  25. Ramadan MA, Tawfik AF, Shibl AM, Gemmell CG. Post-antibiotic effect of azithromycin and erythromycin on streptococcal susceptibility to phagocytosis. J Med Microbiol. 1995;42(5):362ā€“6. https://0-doi-org.brum.beds.ac.uk/10.1099/00222615-42-5-362.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  26. Pfaller MA, Sheehan DJ, Rex JH. Determination of fungicidal activities against yeasts and molds: lessons learned from bactericidal testing and the need for standardization. Clin Microbiol Rev. 2004;17(2):268ā€“80. https://0-doi-org.brum.beds.ac.uk/10.1128/cmr.17.2.268-280.2004.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  27. Simner PJ, Hindler JA, Bhowmick T, Das S, Johnson JK, Lubers BV, Redell MA, Stelling J, Erdman SM. Whatā€™s New in Antibiograms? Updating CLSI M39 Guidance with current trends. J Clin Microbiol. 2022;60(10):e02210ā€“21. https://0-doi-org.brum.beds.ac.uk/10.1128/jcm.02210-21.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  28. Hirshfield I, Barua S, Basu P. Overview of biofilms and some key methods for their study. In: (Goldman E, Green LH, eds) Practical Handbook of Microbiology. 2nd ed. Chapter 42, p.Ā 675ā€“688. Boca Raton: CRC Press; 2009.

  29. Trafny EA, Lewandowski R, Zawistowska-Marciniak I, Stępińska M. Use of MTT assay for determination of the biofilm formation capacity of microorganisms in metalworking fluids. World J Microbiol Biotechnol. 2013;29(9):1635ā€“43. https://0-doi-org.brum.beds.ac.uk/10.1007/s11274-013-1326-0.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  30. Li Q, Yan W, Yang K, Wen Y, Tang JL. Xanthan gum production by Xanthomonas campestris Pv. Campestris 8004 using cassava starch as carbon source. Afr J Biotechnol. 2012;11(73):13809ā€“13. https://0-doi-org.brum.beds.ac.uk/10.5897/AJB11.3774.

    ArticleĀ  CASĀ  Google ScholarĀ 

  31. Lowry OH, Rosebrough NJ, Farr AL, Randall RJ. Protein measurement with the Folin phenol reagent. J Biol Chem. 1951;193:265ā€“75.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  32. Dulley JR, Grieve PA. A simple technique for eliminating interference by detergents in the Lowry method of protein determination. Anal Biochem. 1975;64(1):136ā€“41. https://0-doi-org.brum.beds.ac.uk/10.1016/0003-2697(75)90415-7.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  33. Mishra M, Tiwari S, Gomes AV. Protein purification and analysis: next generation Western blotting techniques. Expert Rev Proteomics. 2017;14(11):1037ā€“53. https://0-doi-org.brum.beds.ac.uk/10.1080/14789450.2017.1388167.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  34. Suzuki J, Kunimoto T, Hori M. Effects of kanamycin on protein synthesis: inhibition of elongation of peptide chains. J Antibiot. 1970;23(2):99ā€“101. https://0-doi-org.brum.beds.ac.uk/10.7164/antibiotics.23.99.

    ArticleĀ  CASĀ  Google ScholarĀ 

  35. Misko TP, Schilling RJ, Salvemini D, Moore WM, Currie MG. A fluorometric assay for the measurement of Nitrite in biological samples. Anal Biochem. 1993;214(1):11ā€“6. https://0-doi-org.brum.beds.ac.uk/10.1006/abio.1993.144.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  36. Auger C, Lemire J, Cecchini D, Bignucolo A, Appanna VD. The metabolic reprogramming evoked by nitrosative stress triggers the anaerobic utilization of citrate in Pseudomonas fluorescens. PLoS ONE. 2011;6(12):e28469. https://0-doi-org.brum.beds.ac.uk/10.1371/journal.pone.0028469.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  37. Andrews S. FastQC: a quality control tool for high throughput sequence data. 2010.

  38. Chen S, Zhou Y, Chen Y, Gu J. Fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics. 2018;34(17):i884ā€“90.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  39. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods. 2012;9(4):357ā€“9.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  40. Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014;30(7):923ā€“30.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  41. Conesa A, Gƶtz S. Blast2GO: a comprehensive suite for functional analysis in plant genomics. Int. J. Plant Genomics. 2008;2008.

  42. Ye J, Zhang Y, Cui H, Liu J, Wu Y, Cheng Y, Xu H, Huang X, Li S, Zhou A, Zhang X. WEGO 2.0: a web tool for analyzing and plotting GO annotations, 2018 update. Nucleic Acids Res. 2018;46(W1):W71ā€“5.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  43. Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, Simonovic M, Doncheva NT, Morris JH, Bork P, Jensen LJ. STRING v11: proteinā€“protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019;47(D1):D607ā€“13. https://0-doi-org.brum.beds.ac.uk/10.1093/nar/gky1131.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  44. Chin CH, Chen SH, Wu HH, Ho CW, Ko MT, Lin CY. cytoHubba: identifying hub objects and sub-networks from complex interactome. BMC Syst Biol. 2014;8(4):1ā€“7. https://0-doi-org.brum.beds.ac.uk/10.1186/1752-0509-8-s4-s11.

    ArticleĀ  Google ScholarĀ 

  45. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498ā€“504. https://0-doi-org.brum.beds.ac.uk/10.1101/gr.1239303.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  46. Untergasser A, Nijveen H, Rao X, Bisseling T, Geurts R, Leunissen JA. Primer3Plus, an enhanced web interface to Primer3. Nucleic Acids Res. 2007;35(suppl2):W71ā€“4.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  47. Calabrese EJ. Hormesis: a fundamental concept in biology. Microb Cell. 2014;1(5):145. https://0-doi-org.brum.beds.ac.uk/10.15698/mic2014.05.145.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  48. Joshi C, Kothari V, Patel P. Importance of selecting appropriate wavelength, while quantifying growth and production of quorum sensing regulated pigments in bacteria. Recent Pat Biotechnol. 2016;10(2):145ā€“52.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  49. Kang D, Kirienko DR, Webster P, Fisher AL, Kirienko NV. Pyoverdine, a siderophore from Pseudomonas aeruginosa, translocates into C. Elegans, removes iron, and activates a distinct host response. Virulence. 2018;9(1):804ā€“17. https://0-doi-org.brum.beds.ac.uk/10.1080/21505594.2018.1449508.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  50. Rada B, Leto TL. Pyocyanin effects on respiratory epithelium: relevance in Pseudomonas aeruginosa airway Infections. Trends Microbiol. 2013;21(2):73ā€“81.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  51. Kasthuri T, Barath S, Nandhakumar M, Karutha Pandian S. Proteomic profiling spotlights the molecular targets and the impact of the natural antivirulent umbelliferone on stress response, virulence factors, and the quorum sensing network of Pseudomonas aeruginosa. Front Cell Infect Microbiol. 2022;12:998540.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  52. Wilson M. An introduction to the human-microbe symbiosis. Microbial inhabitants of humans: their ecology and role in health and Disease. 2nd ed. Cambridge University Press; 2005. p. 15. Chap. 1.

  53. Deforges L, Le Van Thoi J, Soussy CJ, Duval J. Activity of the Amoxicillin-clavulanic acid (augmentin) combination on strains of hospital isolates. Pathol Biol. 1985;33(5):301ā€“8.

    CASĀ  PubMedĀ  Google ScholarĀ 

  54. Khan T, Ullah H, Nasar A, Ullah M. Antibiotic Resistance and sensitivity pattern of Pseudomonas aeruginosa obtained from clinical samples. Lett Appl NanoBioScience. 2023;12(4):112. https://0-doi-org.brum.beds.ac.uk/10.33263/LIANBS124.112.

    ArticleĀ  Google ScholarĀ 

  55. Cervantes-Vega C, ChĆ”vez J, RodrĆ­guez MG. Antibiotic susceptibility of clinical isolates of Pseudomonas aeruginosa. Antonie Van Leeuwenhoek. 1986;52:319ā€“24. https://0-doi-org.brum.beds.ac.uk/10.1007/BF00428643.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  56. Gupta P, Mankere B, Chekkoora Keloth S, Tuteja U, Pandey P, Chelvam KT. Increased antibiotic resistance exhibited by the biofilm of Vibrio cholerae O139. J Antimicrob Chemother. 2018;73(7):1841ā€“7. https://0-doi-org.brum.beds.ac.uk/10.1093/jac/dky127.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  57. Mohammed M, Mekala LP, Chintalapati S, Chintalapati VR. New insights into aniline toxicity: aniline exposure triggers envelope stress and extracellular polymeric substance formation in Rubrivivax benzoatilyticus JA2. J Hazard Mater. 2020;385:121571. https://0-doi-org.brum.beds.ac.uk/10.1016/j.jhazmat.2019.121571.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  58. Oā€™Toole G, Kaplan HB, Kolter R. Biofilm formation as microbial development. Annu Rev Microbiol. 2000;54(1):49ā€“79. https://0-doi-org.brum.beds.ac.uk/10.1146/annurev.micro.54.1.49.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  59. Yang L, Haagensen JA, Jelsbak L, Johansen HK, Sternberg C, HĆøiby N, Molin S. In situ growth rates and biofilm development of Pseudomonas aeruginosa populations in chronic lung Infections. J Bacteriol. 2008;193(9):2760ā€“8. https://0-doi-org.brum.beds.ac.uk/10.1128/jb.01581-07.

    ArticleĀ  Google ScholarĀ 

  60. Hu Y, Wang J, Sun H, Wang S, Liao X, Wang J, An T. Roles of extracellular polymeric substances in the bactericidal effect of nanoscale zero-valent iron: trade-offs between physical disruption and oxidative damage. Env Sci Nano. 2019;6(7):2061ā€“73.

    ArticleĀ  CASĀ  Google ScholarĀ 

  61. Morales DK, Grahl N, Okegbe C, Dietrich LE, Jacobs NJ, Hogan DA. Control of Candida albicans metabolism and biofilm formation by Pseudomonas aeruginosa phenazines. MBio. 2013;4(1):e00526ā€“12. https://0-doi-org.brum.beds.ac.uk/10.1128/mbio.00526-12.

  62. Davies DG, Parsek MR, Pearson JP, Iglewski BH, Costerton JW, Greenberg EP. The involvement of cell-to-cell signals in the development of a bacterial biofilm. Science. 1998;280(5361):295ā€“8. https://0-doi-org.brum.beds.ac.uk/10.1126/science.280.5361.295.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  63. Singh VK, Jayaswal RK, Wilkinson BJ. Cell wall-active antibiotic induced proteins of Staphylococcus aureus identified using a proteomic approach. FEMS Microbiol Lett. 2001;199(1):79ā€“84. https://0-doi-org.brum.beds.ac.uk/10.1111/j.1574-6968.2001.tb10654.x.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  64. Liu B, Qian SB. Translational reprogramming in cellular stress response. Wiley Interdiscip Rev : RNA. 2014;5(3):301ā€“5. https://0-doi-org.brum.beds.ac.uk/10.1002/wrna.1212.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  65. Joshi C, Patel P, Palep H, Kothari V. Validation of the anti-infective potential of a polyherbal ā€˜Panchvalkalā€™ preparation, and elucidation of the molecular basis underlining its efficacy against Pseudomonas aeruginosa. BMC Complement Altern Med. 2019;19(1):1ā€“5. https://0-doi-org.brum.beds.ac.uk/10.1186/s12906-019-2428-5.

    ArticleĀ  CASĀ  Google ScholarĀ 

  66. Ruparel FJ, Shah SK, Patel JH, Thakkar NR, Gajera GN, Kothari VO. Network analysis for identifying potential anti-virulence targets from whole transcriptome of Pseudomonas aeruginosa and Staphylococcus aureus exposed to certain anti-pathogenic polyherbal formulations. Drug Target Insights. 2023;17:58. https://0-doi-org.brum.beds.ac.uk/10.33393/dti.2022.2595.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  67. Poole RK. Nitric oxide and nitrosative stress tolerance in bacteria. Biochem Soc Trans. 2005;33(1):176ā€“80. https://0-doi-org.brum.beds.ac.uk/10.1042/BST0330176.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  68. Heales SJ, BolaƱos JP, Stewart VC, Brookes PS, Land JM, Clark JB. Nitric oxide, mitochondria and neurological Disease. Biochim Biophys Acta - Bioenerg. 1999;1410(2):215ā€“28. https://0-doi-org.brum.beds.ac.uk/10.1016/S0005-2728(98)00168-6.

    ArticleĀ  CASĀ  Google ScholarĀ 

  69. Wesselink E, Koekkoek WA, Grefte S, Witkamp RF, Van Zanten AR. Feeding mitochondria: potential role of nutritional components to improve critical Illness convalescence. Clin Nutr. 2019;38(3):982ā€“95. https://0-doi-org.brum.beds.ac.uk/10.1016/j.clnu.2018.08.032.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  70. Schreiber K, Boes N, Eschbach M, Jaensch L, Wehland J, Bjarnsholt T, Givskov M, Hentzer M, Schobert M. Anaerobic survival of Pseudomonas aeruginosa by pyruvate fermentation requires an usp-type stress protein. J Bacteriol. 2006;188(2):659ā€“68. https://0-doi-org.brum.beds.ac.uk/10.1128/jb.188.2.659-668.2006.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  71. Ciofu O. Pseudomonas aeruginosa chromosomal beta-lactamase in patients with cystic fibrosis and chronic lung Infection. Mechanism of antibiotic resistance and target of the humoral immune response. APMIS Suppl. 2003:(116):1ā€“47.

  72. Litwin CM, Calderwood S. Role of iron in regulation of virulence genes. Clin Microbiol Rev. 1993;6(2):137ā€“49. https://0-doi-org.brum.beds.ac.uk/10.1128/cmr.6.2.137.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  73. Lee YH, Cheng FY, Chiu HW, Tsai JC, Fang CY, Chen CW, Wang YJ. Cytotoxicity, oxidative stress, apoptosis and the autophagic effects of silver nanoparticles in mouse embryonic fibroblasts. Biomaterials. 2014;35(16):4706ā€“15. https://0-doi-org.brum.beds.ac.uk/10.1016/j.biomaterials.2014.02.021.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  74. Ahn JM, Eom HJ, Yang X, Meyer JN, Choi J. Comparative toxicity of silver nanoparticles on oxidative stress and DNA damage in the nematode, Caenorhabditis elegans. Chemosphere. 2014;108:343ā€“52. https://0-doi-org.brum.beds.ac.uk/10.1016/j.chemosphere.2014.01.078.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  75. Hande S, Sonkar V, Bhoj P, Togre N, Goswami K, Dash D. The role of oxidative and nitrosative stress of silver nanoparticles in human parasitic helminth Brugia malayi: a mechanistic insight. Acta Parasitol. 2021;1ā€“0. https://0-doi-org.brum.beds.ac.uk/10.1007/s11686-021-00394-4.

  76. Morones-Ramirez JR, Winkler JA, Spina CS, Collins JJ. Silver enhances antibiotic activity against gram-negative bacteria. Sci Transl Med. 2013;5(190):190ra81. https://0-doi-org.brum.beds.ac.uk/10.1126/scitranslmed.3006276.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  77. McQuillan JS, Shaw AM. Differential gene regulation in the Ag nanoparticle and Ag+-induced silver stress response in Escherichia coli: a full transcriptomic profile. Nanotoxicology. 2014;8(sup1):177ā€“84. https://0-doi-org.brum.beds.ac.uk/10.3109/17435390.2013.870243.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  78. Yin IX, Zhang J, Zhao IS, Mei ML, Li Q, Chu CH. The antibacterial mechanism of silver nanoparticles and its application in dentistry. Int J Nanomed. 2020;2555ā€“62. https://0-doi-org.brum.beds.ac.uk/10.2147/IJN.S246764.

  79. Li WR, Xie XB, Shi QS, Zeng HY, Ou-Yang YS, Chen YB. Antibacterial activity and mechanism of silver nanoparticles on Escherichia coli. Appl Microbiol Biotechnol. 2010;85:1115ā€“22. https://0-doi-org.brum.beds.ac.uk/10.1007/s00253-009-2159-5.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  80. Å inko G, Vinković Vrček I, Goessler W, Leitinger G, DijanoÅ”ić A, Miljanić S. Alteration of cholinesterase activity as possible mechanism of silver nanoparticle toxicity. Environ Sci Pollut Res. 2014;21:1391ā€“400. https://0-doi-org.brum.beds.ac.uk/10.1007/s11356-013-2016-z.

    ArticleĀ  CASĀ  Google ScholarĀ 

  81. Subhadra B, Kim DH, Woo K, Surendran S, Choi CH. Control of biofilm formation in healthcare: recent advances exploiting quorum-sensing interference strategies and multidrug efflux pump inhibitors. Materials. 2018;11(9):1676. https://0-doi-org.brum.beds.ac.uk/10.3390/ma11091676.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  82. Qais FA, Shafiq A, Ahmad I, Husain FM, Khan RA, Hassan I. Green synthesis of silver nanoparticles using Carum copticum: Assessment of its quorum sensing and biofilm inhibitory potential against gram negative bacterial pathogens. Microb Pathog. 2020;144:104172. https://0-doi-org.brum.beds.ac.uk/10.1016/j.micpath.2020.104172.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  83. Feng QL, Wu J, Chen GQ, Cui FZ, Kim TN, Kim JO. A mechanistic study of the antibacterial effect of silver ions on Escherichia coli and Staphylococcus aureus. J Biomed Mater Res. 2000;52(4):662ā€“8. https://0-doi-org.brum.beds.ac.uk/10.1002/1097-4636.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

Download references

Acknowledgements

Authors thank Nirma Education and Research Foundation (NERF), Ahmedabad for infrastructural support, and for providing stipend to NT. GG acknowledges scholarship from Government of Gujarat through their SHODH scheme.

Funding

This research received no extramural funding.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: DM and VK.; Methodology: GG, NT, and VK.; Formal analysis, investigation, data curation: NT, GG.; Resources: DM, VK; Writingā€”original draft preparation: GG, NT, and VK; Writingā€”review and editing: DM, CG, AD; Supervision and project administration: VK; Funding acquisition: DM and VK.

Corresponding author

Correspondence to Vijay Kothari.

Ethics declarations

Ethics approval and consent to participate

Not Applicable.

Consent for publication

Not Applicable.

Competing interests

Three of the authors DM, AD, and CG are from Viridis Biopharma Pvt. Ltd., who manufacture and market SilversolĀ®. However, this has not affected anyway the design of the study or interpretation of data. Rest of the authors have no competing interests to declare.

Additional information

Publisherā€™s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the articleā€™s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the articleā€™s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gajera, G., Thakkar, N., Godse, C. et al. Sub-lethal concentration of a colloidal nanosilver formulation (SilversolĀ®) triggers dysregulation of iron homeostasis and nitrogen metabolism in multidrug resistant Pseudomonas aeruginosa. BMC Microbiol 23, 303 (2023). https://0-doi-org.brum.beds.ac.uk/10.1186/s12866-023-03062-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s12866-023-03062-x

Keywords