AAC Accepted Manuscript Posted Online 27 April 2015 Antimicrob. Agents Chemother. doi:10.1128/AAC.05177-14 Copyright © 2015, American Society for Microbiology. All Rights Reserved.

Silver oxynitrate: An Unexplored Silver Compound with Antimicrobial and Antibiofilm Activity Joe A. Lemire1, Lindsay Kalan2, Alexandru Bradu1, and Raymond J. Turner1,# 1. 2.

The Biofilm Research Group. Department of Biological Sciences at the University of Calgary. 2500 University Drive Northwest, Calgary, AB T2N 1N4 Exciton Technologies Inc. 10230 Jasper Ave NW, Edmonton, AB T5J 4P6

# Corresponding Author – Email: [email protected], Phone: 403-220-4308

Running title: Silver oxynitrate has antimicrobial and antibiofilm efficacy Keywords: silver, antimicrobial, biofilm, antibiofilm, wound care, chronic wound treatment

J.Lemire et al. 2015 1

ABSTRACT:

2

Historically its been accepted, and recent research has established that silver (Ag) is an

3

efficacious antimicrobial agent. A dwindling pipeline of new antibiotics, combined with an increase in

4

antibiotic resistant infections, is bringing Ag to the fore as a therapeutic compound to treat infectious

5

diseases. Currently, many formulations of Ag are being deployed for commercial and medical

6

purposes with varying degrees of effectiveness at killing microbial cells. Here, we evaluated the

7

antimicrobial and antibiofilm capacity of our lead compound – silver oxynitrate [Ag(Ag3O4)2NO3 or

8

Ag7NO11] – against other metal compounds with documented antimicrobial activity; Ag2SO4, AgNO3,

9

silver sulfadiazine (AgSD), AgO, Ag2O, and CuSO4. Our findings reveal that Ag7NO11 eradicates

10

biofilm and planktonic populations, of Pseudomonas aeruginosa, Escherichia coli, Staphylococcus

11

aureus, uropathogenic Escherichia coli (UPEC), fluoroquinolone-resistant Pseudomonas aeruginosa

12

(FQRP), and methicillin-resistant Staphylococcus aureus (MRSA) at lower concentrations than the

13

other tested metal salts. Altogether, our results demonstrate that Ag7NO11 has an enhanced efficacy for

14

the treatment of biofilm-forming pathogens.

15 16 17 18 19 20 21 22 23 24 25 2

J.Lemire et al. 2015 26

INTRODUCTION

27

History has demonstrated that silver (Ag) is an efficacious antimicrobial agent, finding utility as

28

an antiquated preservative for food and water (1, 2). Currently, over 100 Ag-containing medical

29

devices

30

(http://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfpmn/pmn.cfm), and there’s mounting evidence

31

that Ag may be effective for preventing the spread of infectious disease (1, 3, 4). Though the

32

toxicological profile of orally administered Ag remains to be resolved, topical application for the

33

treatment of chronic wounds remains promising (5-8).

34

health issue, as chronic wounds represent a substantial – financial and medical – burden to the health

35

care system. One of the hallmarks of a chronic wound is the presence of a biofilm; a factor that

36

complicates wound healing, and is hypothesized to be the fulcrum between an acute to chronic wound

37

transition (6, 9-15). Also, bacterial biofilms contaminate implanted medical devices (16) and exhibit

38

high resistance to conventional antibiotics (17-23). New antibiofilm agents are thus desperately needed

39

in medicine.

40

have

been

approved

for

use

by

the

FDA

Accordingly, Ag can address a timely public

Recently, researchers have demonstrated that Ag formulations also have potential as antibiofilm

41

agents; this includes AgSD (24), Ag-nanoparticles [AgNPs] (25, 26), and AgNO3 (27). Currently, the

42

wound care industry is replete with Ag-based ointments and wound dressings with claimed efficacy at

43

reducing bacterial bioburden (7, 28-32). However, only a handful of antibiofilm products exist. There

44

are numerous reasons why this is so. 1) Current standardized, antimicrobial testing methods focus on

45

bacteria in their planktonic state – not as biofilms (33, 34). 2) Biofilms are characteristically more

46

tolerant to metal poisoning (35-38), and 3) the detailed mechanism of action for the toxicity of Ag, to

47

biofilms, remains to be fully described (1).

48

The antimicrobial activity of Ag is intrinsically dependent on the formation of the Ag1+ ion (7,

49

39). Briefly, Ag1+ - a lewis soft acid – poisons the microbial cell by binding to reduced thiols (SH),

50

impairing membrane function (1), and disrupting iron-sulfur clusters (40). That being said, the 3

J.Lemire et al. 2015 51

antimicrobial efficacy of higher oxidation states of Ag – Ag2+ and Ag3+ - has not been given adequate

52

consideration, mainly due to instability in solution. A promising resolution to the instability of higher

53

oxidation states of Ag, are silver oxysalts; for example Ag(Ag3O4)2X -the most stable of which is

54

coordinated with nitrate- silver oxynitrate [Ag(Ag3O4)2 NO3 = Ag7NO11], where both the Ag II and III

55

oxidation states exist stably at room temperature (41, 42).

56

Here, we describe our observations from testing the antimicrobial and antibiofilm activity of

57

various metal compounds. Namely, we tested silver oxynitrate (Ag7NO11), silver sulfadiazine (AgSD),

58

silver nitrate (AgNO3), silver sulphate (Ag2SO4), silver (I,III) oxide (AgO), silver (I) oxide (Ag2O), and

59

copper sulphate (CuSO4) against 6 bacterial strains - Pseudomonas aeruginosa (PAO1),

60

Staphylococcus aureus (ATCC 25923), Escherichia coli (JM109), uropathogenic Escherichia coli

61

(CFT703) [UPEC], fluoroquinolone-resistant Pseudomonas aeruginosa [FQRP], and methicillin-

62

resistant Staphylococcus aureus (USA300) [MRSA] as well as two fungal strains of Candida albicans

63

(ATCC 14053) and Candida tropicalis (99916). CuSO4 was included in our analysis because copper

64

(Cu) is another thiophilic metal with observed antimicrobial activity (1, 43, 44). Using the minimal

65

biofilm eradication concentration (MBEC) assay, we found that Ag7NO11 eradicated planktonic and

66

biofilm populations, of the tested strains, at lower concentrations than the other metal compounds

67

tested. AgNO3 – the most soluble Ag salt – also had antimicrobial and antibiofilm activity at

68

micromolar concentrations. Because AgNO3 has a high solubility, resulting in easily accessible Ag+

69

ions, and it outperformed all other Ag salts, with the exception of Ag7NO11, it was chosen as our

70

comparator for testing the efficacy of Ag7NO11 in proceeding experiments. Using the live/dead™

71

staining technique as well as crystal violet staining, we demonstrated that Ag7NO11 reduced the

72

biomass of the biofilm. In summary, our data demonstrates that Ag7NO11 has greater antimicrobial and

73

antibiofilm activity, compared to the other tested metal compounds, and may offer an alternative option

74

for the treatment of chronic wounds infected by biofilm-forming microbes.

75 4

J.Lemire et al. 2015 76

MATERIALS AND METHODS

77

Bacterial/Fungal Strains and Media

78

Bacterial and fungal strains were stored in Microbank ™ vials at -70°C as described by the

79

manufacturer (ProLab Diagnostics, Richmond Hill, ON, Canada). Pseudomonas aeruginosa (PA01),

80

Staphylococcus aureus (ATCC 25923), Escherichia coli (JM109), Candida albicans (ATCC14053),

81

Candida tropicalis (99916), uropathogenic Escherichia coli (CFT703), fluoroquinolone-resistant

82

Pseudomonas aeruginosa, and methicillin-resistant Staphylococcus aureus (USA300) were streak-

83

purified onto tryptic soy agar [TSA] (VWR International, Mississauga, ON, Canada) overnight at 37°C.

84

CFT703 and USA300 were generous gifts from Dr. Joe J. Harrison (the University of Calgary), while

85

the FQRP isolate was a generous gift from Dr. Michael Parkins (the University of Calgary). To

86

simulate a wound environment, simulated wound fluid (SWF) [50% peptone water (0.85% NaCl,

87

0.1g/L peptone):50% fetal calf serum (Invitrogen, Life Technologies)] (31, 45) was used as growth

88

media and for susceptibility testing.

89 90 91

Biofilm Formation All biofilms used in this study were cultivated using the Calgary Biofilm Device

92

(CBD)/MBEC™ Device as previously described in (38, 46-50) and by the manufacturers guidelines

93

(Innovotech, Edmonton, AB, Canada). Briefly, following the overnight growth of the pre-culture, the

94

colonies were suspended in SWF to match the density of a 1.0 McFarland standard. The cultures made

95

with the optical standard were subsequently diluted 30 times in 150μL of SWF, which served as the

96

inoculum for the CBD. The biofilm was formed by placing the lid of the CBD – 96 equivalent pegs –

97

into a 96-well microtitre plate (Nunclon, VWR, International) containing the inoculum. The CBD was

98

then placed on a gyrorotary shaker operating at 150rpm in a humidified incubator at 37°C for 4h or

99

24h. For mature biofilm experiments, the inoculated CBD was incubated for a period of 4d

5

J.Lemire et al. 2015 100

(P.aeruginosa), or 6d (S.aureus and E.coli). A period exceeding 4d resulted in overgrowth of PAO1 in

101

the MBEC™ device (personal observation)

102 103 104

Stock Metal Solutions Silver nitrate (AgNO3), silver sulfadiazine (AgSD), silver sulphate (Ag2SO4), silver (I) oxide

105

(Ag2O), silver (I,III) oxide (AgO), and copper sulphate (CuSO4), were all obtained from Sigma-Aldrich

106

(St. Louis, MO, USA). Silver oxynitrate (Ag7NO11), was obtained from Exciton Technologies Inc

107

(Edmonton, AB, Canada). All stock Ag solutions were made at equivalent molarities of Ag – up to

108

5mM – in distilled and deionized (dd)H2O. The CuSO4 stock was made up to 2M in ddH2O. All metal

109

solutions were stored in glass vials at 21°C for no longer than one week. Working solutions were made

110

in SWF, from stock metal solution, no more than 30 min prior to experimental use. From these, serial-

111

dilutions (dilution factor of 2) were made in 96-well plates (the challenge plate). The first row was

112

reserved as a sterility control and the second row for a growth control (0mM metal salt). The range of

113

concentrations tested for all Ag compound was 0 – 1250μM, while CuSO4 was tested from 0 –

114

20,000μM.

115 116

Biofilm and Planktonic Culture Susceptibility Testing

117

Two scenarios were tested i) the capacity of metal salts to prevent biofilm formation, and ii) the

118

ability of metal salts to eradicate established biofilms. For i) the bacterial cultures were inoculated into

119

the challenge plate in the presence of the metal salts and then subsequently placed on a gyrorotary

120

shaker at 150rpm, in a humidified incubator at 37°C for 4h. Scenario ii) allowed for the establishment

121

of a biofilm for 24h, 4d or 6d on the pegged lid of the CBD. The peg lid containing the established

122

biofilm was then rinsed twice with 0.9% NaCl and subsequently placed into the 96-well microtitre plate

123

containing the two-fold serial dilutions of the challenge metal (the challenge plate). The plate was then

124

incubated for 4h, 24h, or 48h on a gyrorotary shaker at 150rpm, in a humidified incubator at 37°C. 6

J.Lemire et al. 2015 125

To test the metal susceptibility of the planktonic and biofilm populations, the plates from i and

126

ii were prepared by removing the peg lid and rinsing it twice with 0.9% NaCl. The biofilms were

127

disrupted from the pegs by sonication using a 250HT ultrasonic cleaner (VWR, International), set at

128

60Hz for 10min, into 200μL of tryptic soy broth (VWR, International) containing universal neutralizer

129

(UN) [0.05 g/L-Histidine (Sigma, USA), 0.05 g/L-Cysteine (Sigma, USA), and 0.l g/L reduced

130

glutathione (Sigma, USA) ddH2O] and 0.1% Tween® 20. The minimal biofilm eradication

131

concentration (MBEC) of the biofilm populations, was ascertained by performing 8 dilutions -dilution

132

factor of 10- of the disrupted biofilms in 0.9% NaCl. The viable cell numbers from the biofilm were

133

ascertained by spot plating the diluted sample onto TSA plates, and subsequently inoculating the plates

134

overnight at 37°C (38, 51, 52). Similarly, the minimal bactericidal concentration (MBC) of the

135

planktonic populations was carried out by serially diluting, 8 times (10-fold dilutions), the neutralized

136

(UN; as described above) spent media from the 96-well plate into 0.9% saline. Viable cell counts were

137

taken following spot plating of the dilutions onto TSA plates incubated overnight at 37°C (38, 51, 52).

138

The MBC and MBEC were determined by monitoring the concentration of metal compound at which

139

there were no viable microbial colonies.

140 141 142

Confocal Microscopy Quantification of live and dead cells in the biofilm was performed using a confocal microscope

143

and the Live/Dead® BacLight™ staining kit (Molecular Probes, Burlington, ON, Canada). Briefly,

144

following metal exposure, cell viability staining of the bacteria was done by incubating the pegs

145

containing the biofilms with the Syto-9 (6.7μM) and propidium iodide (PI) (40μM) stains (provided in

146

the BacLight™ kit) at 30°C for 30min as previously described in (46). The pegs were rinsed with 0.9%

147

saline twice and monitored using a confocal laser-scanning microscope (Leica DM IRE2 microscope)

148

equipped with a 20X objective. Images were processed using Imaris x64 (Bitplane, USA). PI

149

excitation wavelength was at 523nm and emission at 617nm (red), while Syto-9 excitation wavelength 7

J.Lemire et al. 2015 150

was 488nm and emission at 498 (green). Quantification of the compiled confocal images was

151

performed using the Fiji software package (http://fiji.sc/Fiji).

152 153

Crystal Violet Assay

154

Crystal violet staining was used to quantify the amount of biomass in the biofilm. Following

155

metal treatment, the peg lids from the CBD were rinsed twice in 200μL of 0.9% saline. Following a

156

procedure similar to O’Toole GA, 2011 (53), the biofilms were stained with 200μL of 0.1% crystal

157

violet solution for 10min. Following the staining, the pegs were washed with 200μL ddH2O thrice, and

158

blotted on paper towel to remove excess dye. The stain was left to set overnight at room temperature.

159

Quantification of the biofilm was performed by solubilizing the crystal violet stain with 200μL of 30%

160

acetic acid for 15min, into a microtitre plate, and reading the absorbance at 550nm using acetic acid as

161

the blank.

162 163 164

Statistical Analysis Statistical significance of the results was performed using a non-parametric, one-way analysis

165

of variance (ANOVA) for the MBC and MBEC assays, a two-way ANOVA for the crystal violet

166

assays, or a students T-test for Supplementary Figure 3 and 4. All experiments were performed, at

167

minimum (stated otherwise in the figure captions), with two biological replicates and in duplicate.

168 169

RESULTS

170

Silver oxynitrate (Ag7NO11) prevents biofilm formation. In order to assess the capacity of various

171

metal compounds to prevent biofilm formation in our selected pathogenic strains (E.coli, P.aeruginosa,

172

and S.aureus), we employed the minimal biofilm eradication concentration (MBEC) assay (Fig. 1).

173

Concurrently, the minimal bactericidal concentration (MBC) for the metal compounds was also

174

established. Compared to the other metal compounds tested, significantly lower concentrations of 8

J.Lemire et al. 2015 175

Ag7NO11 were needed to prevent the formation of biofilms (Fig. 1A) and –with the exception of

176

AgNO3- eliminate planktonic populations of our tested strains (Fig. 1B), following a 4h incubation

177

period.

178 179

Silver oxynitrate (Ag7NO11) eradicated established biofilm populations. Our next aim was to

180

assess the capacity of various metal compounds to eradicate an established biofilm. Biofilms from

181

E.coli, P.aeruginosa, and S.aureus were established following a 24h incubation period in the MBEC™

182

device. The established biofilms were then exposed to serial dilutions (two-fold) of Ag2O, AgO,

183

Ag2SO4, AgNO3, Ag7NO11, AgSD, and CuSO4 for 24h (Fig. 2). Exposing established biofilms for 4h

184

to AgNO3, Ag7NO11, and CuSO4 was insufficient time to eradicate them (Suppl. Fig. 1) – with the

185

exception of E.coli biofilms. Thus, a 24h exposure time to the metal compounds was evaluated as a

186

potentially efficacious treatment period. Following a 24h incubation period with the various metal

187

compounds, we observed that Ag7NO11 significantly reduced biofilm populations of E. coli, P.

188

aeruginosa, and S. aureus at lower concentrations than the other tested metal compounds – with the

189

exception of AgNO3 in P. aeruginosa (Fig. 2). When mature biofilms were established in the

190

MBEC™ device, following a 4d (P.aeruginosa) or 6d (E.coli and S.aureus), higher concentrations of

191

AgNO3, CuSO4, and Ag7NO11 were required to reach a MBEC with the exception of CuSO4 in E.coli

192

(Fig. 3). Nonetheless, Ag7NO11 eradicated the mature biofilms of S.aureus and E.coli at lower

193

concentrations than AgNO3 and CuSO4 (Fig. 3). Additionally, at the highest concentrations –of

194

equivalent Ag- of AgNO3 and Ag7NO11 tested (1250μM), Ag7NO11 significantly outperformed AgNO3

195

and CuSO4 at eradicating C.albicans and C.tropicallis biofilms (Suppl. Fig 2).

196 197

Silver oxynitrate (Ag7NO11) reduces the biomass of established biofilms. Since AgNO3 and

198

Ag7NO11 were the most efficacious at eliminating planktonic populations, preventing biofilm

199

formation, and eradicating established biofilms (Fig 1. and Fig. 2), these two metal compounds were 9

J.Lemire et al. 2015 200

chosen as lead compounds to study the capacity of Ag to abolish established biofilms. We employed

201

Live/Dead® staining followed by confocal microscopy analysis to visualize the effects of a 4h and 24h

202

treatment with 5μM and 12.5μM of AgNO3 and Ag7NO11 (at equivalent Ag concentrations) on

203

established biofilms of E. coli, P. aeruginosa, and S. aureus (Suppl. Fig. 3 and Fig. 4). Though, a 4h

204

exposure to AgNO3 and Ag7NO11 was not adequate to eradicate the biofilm populations, an increase in

205

the quantity of dead cells was observed in the 12.5μM AgNO3 and Ag7NO11 – exposed biofilms

206

(Suppl. Fig.3). Following a 24h exposure to AgNO3 and Ag7NO11 we observed an increase in dead

207

cells in the 12.5μM AgNO3-exposed biofilms and reduction in biofilm quantity in the Ag7NO11-

208

exposed biofilms (Fig. 4 and Suppl. Fig. 4). To confirm that Ag7NO11 bestowed the capacity to reduce

209

the quantity of the biomass in the biofilms, we performed a crystal violet staining assay –following the

210

O’Toole method (53) - on the established E .coli, P. aeruginosa, and S .aureus biofilms following a

211

24h exposure to AgNO3, Ag7NO11, and CuSO4 (Fig. 5). We observed that Ag7NO11 had significantly

212

greater efficacy than AgNO3 and CuSO4 at reducing the quantity of biomass in E. coli and P.

213

aeruginosa biofilms – albeit, much higher concentrations were needed to reduce the P. aeruginosa

214

biofilm-biomass (Fig. 5). Meanwhile, Ag7NO11 only outperformed CuSO4 for reducing the biomass of

215

S.aureus biofilms (Fig. 5).

216 217

Silver oxynitrate (Ag7NO11) is more efficacious against select clinical isolates and antibiotic-

218

resistant strains of E.coli [UPEC (CFT703)], P.aeruginosa [FQRP], and S.aureus [MRSA

219

(USA300)]. To demonstrate the potential of using Ag7NO11 as an antimicrobial beyond lab strains of

220

bacterial pathogens, we performed MBC and MBEC assays on UPEC, FQRP, and MRSA planktonic

221

and biofilm populations, respectively, with serial dilutions of Ag7NO11, AgNO3, and CuSO4 (Fig. 6, 7,

222

and 8; respectively). Our observations suggest that greater concentrations of AgNO3, Ag7NO11, and

223

CuSO4 are required to eradicate planktonic and biofilm populations of UPEC, FQRP, and MRSA (Fig.

224

6,7, and 8), with the one exception of Ag7NO11 in FQRP; which has a similar effectiveness at 10

J.Lemire et al. 2015 225

eradicating planktonic populations, as well as preventing and eradicating biofilm populations as it does

226

in the PAO1 strain (Fig. 7 and 1; respectively). Nonetheless, we observed that Ag7NO11 eliminated

227

planktonic populations, prevented biofilm formation, and eradicated established biofilms of UPEC,

228

FQRP, and MRSA at lower concentrations than AgNO3, and CuSO4; with the exception of established

229

UPEC biofilms which had no considerable log-reduction values at the concentrations and treatment

230

times tested (Fig. 6,7,8).

231 232 233

DISCUSSION Biofilms represent an earnest concern in medicine, as they contribute to chronic infections,

234

complicate wound healing, contaminate medical devices, and exhibit resistance to conventional

235

antimicrobials (54-63). Ionic silver (Ag+) has a demonstrated antimicrobial outcome at low

236

concentrations (64-66) (67) [and references within] and may offer an alternative option for treating

237

antibiotic-resistant infections. Here, we demonstrated how silver oxynitrate (Ag7NO11) – a high

238

oxidation state Ag compound - carries an enhanced antimicrobial and antibiofilm capacity.

239

Additionally, our present study adds to the body of literature supporting the antimicrobial and

240

antibiofilm potential of Ag.

241

In this study we confirmed that Ag is effective at eliminating planktonic populations of E. coli,

242

P. aeruginosa, and S. aureus. Additionally, we established that Ag is effective at preventing biofilm

243

formation (Fig. 1) and eradicating established biofilms (Fig. 2). Indeed, we also observed that Cu is

244

effective at killing planktonic and biofilm populations of lab pathogens, albeit at millimolar

245

concentrations. Mechanistically, both Cu and Ag are thiophilic metals that have similar targets in the

246

microbial cell (1). One key distinction between the two metals, is that Cu is an essential metal, while

247

Ag is non-essential and has no known function in the bacterial cell (1). However, it’s important to

248

recognize that the precise manner with which Ag poisons the microbial cell remains to be fully

249

elaborated. 11

J.Lemire et al. 2015 250

One novel highlight of this study was that it demonstrated, for the first time, that Ag7NO11 –a

251

source of higher oxidation states of Ag- had a MBC and MBEC that was much lower (2 fold or lower)

252

than the other metal compounds tested. The advantage of having a lower MBC and MBEC is that less

253

of the compound would be required to have the desired therapeutic effect. Interestingly, Ag7NO11 was

254

not significantly better than AgNO3 at preventing E. coli and P. aeruginosa biofilm formation, or at

255

eradicating established P. aeruginosa biofilms (Fig. 1a and 2). But, Ag7NO11 was significantly better

256

than AgNO3 at eradicating planktonic populations of all tested bacterial strains (Fig. 1b). This

257

observation is not surprising as biofilms are notoriously more difficult to eliminate than their

258

planktonic counterparts (51, 52, 68, 69) – an observation confirmed by this study (Fig. 1, 2, and

259

Suppl. Fig. 1). In terms of the MBC and MBEC of the Ag salts, the trend in efficacy appeared to

260

follow the molar solubility of the salts in water; AgNO3 > Ag2SO4 > AgO > Ag2O (Ksp = 51.6M,

261

1.4x10-5M, 4.8x10-8M, and 3.6x10-11M; respectively). However, we did note strain specific

262

sensitivities. For example, E. coli planktonic populations were more sensitive to Ag2SO4 in

263

comparison to P. aeruginosa and S. aureus. We were surprised at the poor efficacy of AgSD –the

264

silver/sulfonamide synergistic biocidal compound- as it is part of the current standard-of-care for the

265

treatment of burn wounds (70-72). In some cases, CuSO4 outperformed AgSD as a bactericidal

266

compound (Fig. 1B and 2). Not surprisingly, much higher concentrations and treatment periods with

267

Ag and Cu were needed to reduce the viable cells in mature biofilms (4d; P.aeruginosa, or 6d; E.coli

268

and S.aureus) [Fig. 3]. The enhanced metal resistance of mature biofilms has been reported previously

269

(73). However, making comparisons across studies is difficult due to differing methodologies used for

270

growing the biofilms. Regardless, this study demonstrated that Ag7NO11 outperformed AgNO3 and

271

CuSO4 for eradicating mature E.coli and S.aurues biofilms.

272

A universally recognized challenge in treating biofilm-associated infections is reducing the

273

biomass of the biofilm. Without reduction of the biofilm-biomass, many antimicrobials will kill the

274

outermost bacteria in the biofilm, but will fail to penetrate and kill the innermost bacterial cells – 12

J.Lemire et al. 2015 275

resulting in recurring or persistent infections (9, 74-76). Performing live/dead™ staining, following a

276

24h AgNO3 and Ag7NO11 exposure to established biofilms, revealed that Ag7NO11 has the capacity, to

277

reduce the biomass E. coli, P. aeruginosa, and S. aureus biofilms. Both, AgNO3 and Ag7NO11

278

increased the amount of dead cells within the biofilm, but Ag7NO11 was superior for reducing the

279

biomass of the biofilm (Fig. 4 and Suppl. Fig 4). Monitoring the quantity of biomass in established

280

biofilms of E. coli, P. aeruginosa, and S. aureus following a 24h exposure to CuSO4, AgNO3, and

281

Ag7NO11, revealed that Ag7NO11 reduces the amount of biofilm-biomass significantly more than

282

AgNO3 and CuSO4 in E. coli, and P. aeruginosa. Meanwhile, Ag7NO11 was only significantly better

283

than CuSO4 in reducing the biomass of S. aureus biofilms (Fig 4). Interestingly, while the biomass-

284

reducing effects of the metal compounds on E. coli biofilms were concentration dependent, this was not

285

the case with P. aeruginosa or S. aureus. This phenomenon raises interesting insights into the potential

286

necessity to target therapies toward particular infections. However, no conclusions should be drawn

287

from these limited observations.

288

Our overarching goal was to demonstrate the universal antimicrobial and antibiofilm potential

289

of Ag7NO11. Accordingly, we performed an MBC and MBEC assays on a three clinically-relevant

290

pathogenic strains – uropathogenic E.coli, fluoroquinolone-resistant P.aeruginosa, and methicillin-

291

resistant S.aureus - (Fig. 6, 7, and 8) as well as MBEC assays on two laboratory strains of fungi – C.

292

albicans and C. tropicalis (Suppl. Fig. 2). Again, Ag7NO11 had a lower MBC against planktonic

293

populations, prevented biofilm formation, and eradicated established biofilms at lower concentrations

294

when compared to AgNO3 and CuSO4. Certainly, Ag has been explored as a strategy to control

295

infection of uropathogens (77), as well as combatting and controlling antibiotic-resistant pathogens,

296

including MRSA (78-82). The results from this study demonstrated variable sensitivities of the

297

planktonic and biofilm populations to CuSO4, AgNO3, and Ag7NO11. In general, all of the planktonic

298

and biofilm populations of the clinically-relevant strains were capable of tolerating higher

299

concentrations of the tested metals than their laboratory counterparts (Fig. 1, 2, 6, 7, and 8). The single 13

J.Lemire et al. 2015 300

exception to this trend was observed with FQRP, where Ag7NO11 was exceptionally efficacious in the

301

low micromollar range; tempting speculation that Ag7NO11 can be considered as an antimicrobial

302

coating for endotracheal tubes. It is also noteworthy that Ag7NO11 eliminated planktonic populations,

303

prevented biofilm formation, and eradicated established biofilms at lower concentrations than AgNO3

304

and CuSO4, with the exception of established UPEC biofilms, which could not be reduced to a

305

significant level by Ag under the conditions tested. Again, confirming the challenge involved in

306

eradicating an established biofilm. Though only limited analyses were performed in this study, we

307

observed that Candida sp. biofilms are also difficult to eradicate (Suppl. Fig 4). The enhanced

308

tolerance of Candida sp. has been observed previously within our research group (38, 83, 84).

309

However, Ag7NO11 still has an MBEC in the millimolar range – much within a potentially therapeutic

310

range. This is ever important as Candida sp. are biofilm-forming microorganisms with potential to be

311

infectious and can cause deleterious health outcomes (85-88).

312

To conclude, here we have demonstrated that thiophilic metals – such as Cu and Ag – are

313

efficacious antimicrobial and antibiofilm agents. Furthermore, we’ve demonstrated that silver

314

oxynitrate (Ag7NO11) – a compound that delivers higher oxidations states of Ag – has a greater

315

antimicrobial and antibiofilm capacity, at equal concentrations of Ag, than AgNO3, Ag2SO4, AgO,

316

Ag2O, and CuSO4. Additionally, we demonstrated that Ag7NO11 reduces the biomass of the biofilm –

317

an important strategy for combatting biofilm-associated infections.

318

Recent realizations have focused on biofilms as the causative agent of many infectious diseases,

319

but we are at a loss for adequate therapies to eradicate them. This includes chronic wounds, where

320

many wound care products exist, but very few have an antibiofilm capacity. Fortunately, wound care

321

products offer the greatest potential for implementing metal-based antimicrobials, due to decreased

322

internalization of the therapeutic by the patient. This is the venue with which we envision the best

323

utility for Ag7NO11 as a strategy for preventing and controlling infectious disease. Thus, Ag7NO11 will

14

J.Lemire et al. 2015 324

add to our arsenal for combatting infection, an arsenal that is desperately needed in the face of

325

increasing antibiotic resistant infections and a dwindling pipeline of new antibiotics.

326 327 328

ACKNOWLEDGEMENTS AND CONFLICTS OF INTEREST We would like to extend a special thanks to Dr. Joe Harrison at the University of Calgary for

329

providing the MRSA strain (USA 300) and the uropathogenic E.coli strain (CFT703). We would also

330

like to extend our gratitude to Dr. Michael Parkins at the Cummings School of Medicine for providing

331

us with the fluoroquinolone-resistant P.aeruginosa isolate. J.L. is currently funded by the Natural

332

Sciences and Engineering Council of Canada - Banting Postdoctoral Fellowship Program, as well as

333

Alberta Innovates Health Solutions. A.B. received summer internship support from NSERC. This

334

work was supported by an NSERC-Engage grant and by a MITACS Accelerate fellowship for J.L.;

335

both of which required financial and in-kind contributions from Exciton Technologies Inc. (ETI). L.K

336

was funded by the NSERC Industrial R&D Fellowship (IRDF) and Alberta Innovates Technology

337

Futures (AITF) R&D Associates Fellowship for this work. ETI manufactured and supplied the silver

338

oxynitrate. In addition, ETI provided technical assistance in performing some of the experiments.

339 340 341 342 343 344 345 346 347 348 15

J.Lemire et al. 2015 349

LITERATURE CITED

350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393

1. 2. 3.

4. 5.

6. 7. 8.

9. 10. 11.

12.

13.

14.

15. 16.

17. 18.

Lemire JA, Harrison JJ, Turner RJ. 2013. Antimicrobial activity of metals: mechanisms, molecular targets and applications. Nat Rev Microbiol 11:371-384. Alexander JW. 2009. History of the medical use of silver. Surgical infections 10:289-292. Mijnendonckx K, Leys N, Mahillon J, Silver S, Van Houdt R. 2013. Antimicrobial silver: uses, toxicity and potential for resistance. Biometals : an international journal on the role of metal ions in biology, biochemistry, and medicine 26:609-621. Morones-Ramirez JR, Winkler JA, Spina CS, Collins JJ. 2013. Silver enhances antibiotic activity against gram-negative bacteria. Science translational medicine 5:190ra181. Percival SL, Thomas J, Linton S, Okel T, Corum L, Slone W. 2012. The antimicrobial efficacy of silver on antibiotic-resistant bacteria isolated from burn wounds. International Wound Journal 9:488-493. Toy LW, Macera L. 2011. Evidence-based review of silver dressing use on chronic wounds. Journal of the American Academy of Nurse Practitioners 23:183-192. Walker M, Parsons D. 2014. The biological fate of silver ions following the use of silvercontaining wound care products - a review. Int Wound J 11:496-504. Aziz Z, Abu SF, Chong NJ. 2012. A systematic review of silver-containing dressings and topical silver agents (used with dressings) for burn wounds. Burns : journal of the International Society for Burn Injuries 38:307-318. Costerton JW, Stewart PS, Greenberg EP. 1999. Bacterial biofilms: a common cause of persistent infections. Science 284:1318-1322. James GA, Swogger E, Wolcott R, Pulcini E, Secor P, Sestrich J, Costerton JW, Stewart PS. 2008. Biofilms in chronic wounds. Wound repair and regeneration. 16:37-44. Bjarnsholt T, Kirketerp-Moller K, Jensen PO, Madsen KG, Phipps R, Krogfelt K, Hoiby N, Givskov M. 2008. Why chronic wounds will not heal: a novel hypothesis. Wound repair and regeneration. 16:2-10. Wolcott RD, Rhoads DD, Bennett ME, Wolcott BM, Gogokhia L, Costerton JW, Dowd SE. 2010. Chronic wounds and the medical biofilm paradigm. Journal of wound care 19:45-46, 48-50, 52-43. Percival SL, Hill KE, Williams DW, Hooper SJ, Thomas DW, Costerton JW. 2012. A review of the scientific evidence for biofilms in wounds. Wound Repair and Regeneration 20:647-657. Valente PMDS, Deva A, Ngo Q, Vickery K. 2014. The increased killing of biofilms in vitro by combining topical silver dressings with topical negative pressure in chronic wounds. International Wound Journal:n/a-n/a. Metcalf DG, Bowler PG. 2014. Clinician perceptions of wound biofilm. International Wound Journal:n/a-n/a. Hoiby N, Ciofu O, Johansen HK, Song ZJ, Moser C, Jensen PO, Molin S, Givskov M, Tolker-Nielsen T, Bjarnsholt T. 2011. The clinical impact of bacterial biofilms. International journal of oral science 3:55-65. Mah TF, O'Toole GA. 2001. Mechanisms of biofilm resistance to antimicrobial agents. Trends in microbiology 9:34-39. Lopes SP, Ceri H, Azevedo NF, Pereira MO. 2012. Antibiotic resistance of mixed biofilms in cystic fibrosis: impact of emerging microorganisms on treatment of infection. International journal of antimicrobial agents 40:260-263.

16

J.Lemire et al. 2015 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440

19.

20. 21. 22. 23. 24.

25.

26. 27.

28.

29.

30. 31.

32. 33.

34. 35. 36. 37.

Ito A, Taniuchi A, May T, Kawata K, Okabe S. 2009. Increased antibiotic resistance of Escherichia coli in mature biofilms. Applied and environmental microbiology 75:40934100. Hoiby N, Bjarnsholt T, Givskov M, Molin S, Ciofu O. 2010. Antibiotic resistance of bacterial biofilms. International journal of antimicrobial agents 35:322-332. Stewart PS, Costerton JW. 2001. Antibiotic resistance of bacteria in biofilms. Lancet 358:135-138. Ceri H, Olson ME, Turner RJ. 2010. Needed, new paradigms in antibiotic development. Expert opinion on pharmacotherapy 11:1233-1237. Mah TF, Pitts B, Pellock B, Walker GC, Stewart PS, O'Toole GA. 2003. A genetic basis for Pseudomonas aeruginosa biofilm antibiotic resistance. Nature 426:306-310. Bjarnsholt T, Kirketerp-Moller K, Kristiansen S, Phipps R, Nielsen AK, Jensen PO, Hoiby N, Givskov M. 2007. Silver against Pseudomonas aeruginosa biofilms. APMIS : acta pathologica, microbiologica, et immunologica Scandinavica 115:921-928. Martinez-Gutierrez F, Boegli L, Agostinho A, Sanchez EM, Bach H, Ruiz F, James G. 2013. Anti-biofilm activity of silver nanoparticles against different microorganisms. Biofouling 29:651-660. Fabrega J, Renshaw JC, Lead JR. 2009. Interactions of Silver Nanoparticles with Pseudomonas putida Biofilms. Environmental Science & Technology 43:9004-9009. Monteiro DR, Gorup LF, Silva S, Negri M, de Camargo ER, Oliveira R, Barbosa DB, Henriques M. 2011. Silver colloidal nanoparticles: antifungal effect against adhered cells and biofilms of Candida albicans and Candida glabrata. Biofouling 27:711-719. Herron M, Agarwal A, Kierski PR, Calderon DF, Teixeira LB, Schurr MJ, Murphy CJ, Czuprynski CJ, McAnulty JF, Abbott NL. 2014. Reduction in wound bioburden using a silver-loaded dissolvable microfilm construct. Advanced healthcare materials 3:916-928. Hooper SJ, Percival SL, Hill KE, Thomas DW, Hayes AJ, Williams DW. 2012. The visualisation and speed of kill of wound isolates on a silver alginate dressing. International Wound Journal 9:633-642. Leaper DJ. 2006. Silver dressings: their role in wound management. International Wound Journal 3:282-294. Jones SA, Bowler PG, Walker M, Parsons D. 2004. Controlling wound bioburden with a novel silver-containing Hydrofiber® dressing. Wound Repair and Regeneration 12:288294. Wilkinson LJ, White RJ, Chipman JK. 2011. Silver and nanoparticles of silver in wound dressings: a review of efficacy and safety. Journal of wound care 20:543-549. Baker B, McKernan P, Marsik F. 2014. Clinical and Regulatory Development of Antibiofilm Drugs: The Need, the Potential, and the Challenges, p. 469-486. In Rumbaugh KP, Ahmad I (ed.), Antibiofilm Agents, vol. 8. Springer Berlin Heidelberg. Beloin C, Renard S, Ghigo J-M, Lebeaux D. 2014. Novel approaches to combat bacterial biofilms. Current Opinion in Pharmacology 18:61-68. Chien C-C, Lin B-C, Wu C-H. 2013. Biofilm formation and heavy metal resistance by an environmental Pseudomonas sp. Biochemical Engineering Journal 78:132-137. Harrison JJ, Ceri H, Turner RJ. 2007. Multimetal resistance and tolerance in microbial biofilms. Nat Rev Micro 5:928-938. Harrison JJ, Turner RJ, Ceri H. 2005. Persister cells, the biofilm matrix and tolerance to metal cations in biofilm and planktonic Pseudomonas aeruginosa. Environmental microbiology 7:981-994. 17

J.Lemire et al. 2015 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487

38. 39. 40.

41. 42. 43. 44. 45.

46.

47.

48.

49. 50. 51. 52. 53. 54. 55. 56. 57. 58.

Harrison JJ, Rabiei M, Turner RJ, Badry EA, Sproule KM, Ceri H. 2006. Metal resistance in Candida biofilms. FEMS microbiology ecology 55:479-491. Lansdown AB. 2006. Silver in health care: antimicrobial effects and safety in use. Current problems in dermatology 33:17-34. Xu FF, Imlay JA. 2012. Silver(I), mercury(II), cadmium(II), and zinc(II) target exposed enzymic iron-sulfur clusters when they toxify Escherichia coli. Applied and environmental microbiology 78:3614-3621. McMillan JA. 1962. Higher Oxidation States of Silver. Chemical Reviews 62:65-80. Djokić SS. 2004. Deposition of Silver Oxysalts and Their Antimicrobial Properties. Journal of The Electrochemical Society 151:C359-C364. Ruparelia JP, Chatterjee AK, Duttagupta SP, Mukherji S. 2008. Strain specificity in antimicrobial activity of silver and copper nanoparticles. Acta biomaterialia 4:707-716. Grass G, Rensing C, Solioz M. 2011. Metallic copper as an antimicrobial surface. Applied and environmental microbiology 77:1541-1547. Said J, Dodoo CC, Walker M, Parsons D, Stapleton P, Beezer AE, Gaisford S. 2014. An in vitro test of the efficacy of silver-containing wound dressings against Staphylococcus aureus and Pseudomonas aeruginosa in simulated wound fluid. International journal of pharmaceutics 462:123-128. Harrison JJ, Ceri H, Yerly J, Stremick CA, Hu Y, Martinuzzi R, Turner RJ. 2006. The use of microscopy and three-dimensional visualization to evaluate the structure of microbial biofilms cultivated in the Calgary Biofilm Device. Biological procedures online 8:194-215. Ceri H, Olson ME, Stremick C, Read RR, Morck D, Buret A. 1999. The Calgary Biofilm Device: new technology for rapid determination of antibiotic susceptibilities of bacterial biofilms. Journal of clinical microbiology 37:1771-1776. Olson ME, Ceri H, Morck DW, Buret AG, Read RR. 2002. Biofilm bacteria: formation and comparative susceptibility to antibiotics. Canadian journal of veterinary research = Revue canadienne de recherche veterinaire 66:86-92. Harrison JJ, Ceri H, Roper NJ, Badry EA, Sproule KM, Turner RJ. 2005. Persister cells mediate tolerance to metal oxyanions in Escherichia coli. Microbiology 151:3181-3195. Harrison JJ, Turner RJ, Ceri H. 2005. High-throughput metal susceptibility testing of microbial biofilms. BMC microbiology 5:53. Harrison JJ, Ceri H, Turner RJ. 2007. Multimetal resistance and tolerance in microbial biofilms. Nature reviews. Microbiology 5:928-938. Harrison JJ, Ceri H, Stremick CA, Turner RJ. 2004. Biofilm susceptibility to metal toxicity. Environmental microbiology 6:1220-1227. O'Toole GA. 2011. Microtiter Dish Biofilm Formation Assay. Journal of Visualized Experiments : JoVE:2437. Costerton JW, Stewart PS, Greenberg EP. 1999. Bacterial Biofilms: A Common Cause of Persistent Infections. Science 284:1318-1322. Stewart PS, William Costerton J. Antibiotic resistance of bacteria in biofilms. The Lancet 358:135-138. Donlan RM, Costerton JW. 2002. Biofilms: Survival Mechanisms of Clinically Relevant Microorganisms. Clinical Microbiology Reviews 15:167-193. Høiby N, Bjarnsholt T, Givskov M, Molin S, Ciofu O. Antibiotic resistance of bacterial biofilms. International journal of antimicrobial agents 35:322-332. Marsh PD, Moter A, Devine DA. 2011. Dental plaque biofilms: communities, conflict and control. Periodontology 2000 55:16-35. 18

J.Lemire et al. 2015 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535

59. 60.

61.

62. 63.

64.

65. 66.

67. 68. 69. 70.

71. 72.

73. 74. 75. 76.

Archer NK, Mazaitis MJ, Costerton JW, Leid JG, Powers ME, Shirtliff ME. 2011. Staphylococcus aureus biofilms. Virulence 2:445-459. Høiby N, Ciofu O, Johansen HK, Song Z-j, Moser C, Jensen PØ, Molin S, Givskov M, Tolker-Nielsen T, Bjarnsholt T. 2011. The clinical impact of bacterial biofilms. International journal of oral science 3:55-65. Gurjala AN, Geringer MR, Seth AK, Hong SJ, Smeltzer MS, Galiano RD, Leung KP, Mustoe TA. 2011. Development of a novel, highly quantitative in vivo model for the study of biofilm-impaired cutaneous wound healing. Wound repair and regeneration : official publication of the Wound Healing Society [and] the European Tissue Repair Society 19:400-410. Guo S, Dipietro LA. 2010. Factors affecting wound healing. Journal of dental research 89:219-229. Roy S, Elgharably H, Sinha M, Ganesh K, Chaney S, Mann E, Miller C, Khanna S, Bergdall VK, Powell HM, Cook CH, Gordillo GM, Wozniak DJ, Sen CK. 2014. Mixedspecies biofilm compromises wound healing by disrupting epidermal barrier function. The Journal of pathology 233:331-343. Kim JS, Kuk E, Yu KN, Kim JH, Park SJ, Lee HJ, Kim SH, Park YK, Park YH, Hwang CY, Kim YK, Lee YS, Jeong DH, Cho MH. 2007. Antimicrobial effects of silver nanoparticles. Nanomedicine : nanotechnology, biology, and medicine 3:95-101. Spacciapoli P, Buxton D, Rothstein D, Friden P. 2001. Antimicrobial activity of silver nitrate against periodontal pathogens. Journal of Periodontal Research 36:108-113. Holt KB, Bard AJ. 2005. Interaction of Silver(I) Ions with the Respiratory Chain of Escherichia coli:  An Electrochemical and Scanning Electrochemical Microscopy Study of the Antimicrobial Mechanism of Micromolar Ag+†. Biochemistry 44:13214-13223. Chernousova S, Epple M. 2013. Silver as antibacterial agent: ion, nanoparticle, and metal. Angewandte Chemie 52:1636-1653. Drenkard E. 2003. Antimicrobial resistance of Pseudomonas aeruginosa biofilms. Microbes and infection / Institut Pasteur 5:1213-1219. Anderson GG, O'Toole GA. 2008. Innate and induced resistance mechanisms of bacterial biofilms. Current topics in microbiology and immunology 322:85-105. Atiyeh BS, Costagliola M, Hayek SN, Dibo SA. 2007. Effect of silver on burn wound infection control and healing: review of the literature. Burns : journal of the International Society for Burn Injuries 33:139-148. Fuller FW. 2009. The side effects of silver sulfadiazine. Journal of burn care & research : official publication of the American Burn Association 30:464-470. Ulkur E, Oncul O, Karagoz H, Yeniz E, Celikoz B. 2005. Comparison of silver-coated dressing (Acticoat), chlorhexidine acetate 0.5% (Bactigrass), and fusidic acid 2% (Fucidin) for topical antibacterial effect in methicillin-resistant Staphylococci-contaminated, fullskin thickness rat burn wounds. Burns : journal of the International Society for Burn Injuries 31:874-877. Teitzel GM, Parsek MR. 2003. Heavy metal resistance of biofilm and planktonic Pseudomonas aeruginosa. Applied and environmental microbiology 69:2313-2320. Hall-Stoodley L, Stoodley P. 2009. Evolving concepts in biofilm infections. Cellular microbiology 11:1034-1043. Fux CA, Costerton JW, Stewart PS, Stoodley P. 2005. Survival strategies of infectious biofilms. Trends in microbiology 13:34-40. Stewart PS. 2002. Mechanisms of antibiotic resistance in bacterial biofilms. International journal of medical microbiology : IJMM 292:107-113. 19

J.Lemire et al. 2015 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572

77.

78.

79.

80.

81.

82.

83.

84.

85.

86.

87. 88.

Desai DG, Liao KS, Cevallos ME, Trautner BW. 2010. Silver or Nitrofurazone Impregnation of Urinary Catheters Has a Minimal Effect on Uropathogen Adherence. The Journal of Urology 184:2565-2571. Wright JB, Lam K, Burrell RE. 1998. Wound management in an era of increasing bacterial antibiotic resistance: A role for topical silver treatment. American journal of infection control 26:572-577. Ülkür E, Oncul O, Karagoz H, Yeniz E, Çeliköz B. 2005. Comparison of silver-coated dressing (Acticoat™), chlorhexidine acetate 0.5% (Bactigrass®), and fusidic acid 2% (Fucidin®) for topical antibacterial effect in methicillin-resistant Staphylococcicontaminated, full-skin thickness rat burn wounds. Burns : journal of the International Society for Burn Injuries 31:874-877. Nanda A, Saravanan M. 2009. Biosynthesis of silver nanoparticles from Staphylococcus aureus and its antimicrobial activity against MRSA and MRSE. Nanomedicine: Nanotechnology, Biology and Medicine 5:452-456. Strohal R, Schelling M, Takacs M, Jurecka W, Gruber U, Offner F. 2005. Nanocrystalline silver dressings as an efficient anti-MRSA barrier: a new solution to an increasing problem. Journal of Hospital Infection 60:226-230. Percival SL, Bowler PG, Dolman J. 2007. Antimicrobial activity of silver-containing dressings on wound microorganisms using an in vitro biofilm model. International Wound Journal 4:186-191. Harrison JJ, Turner RJ, Ceri H. 2007. A subpopulation of Candida albicans and Candida tropicalis biofilm cells are highly tolerant to chelating agents. FEMS microbiology letters 272:172-181. Harrison JJ, Ceri H, Yerly J, Rabiei M, Hu Y, Martinuzzi R, Turner RJ. 2007. Metal ions may suppress or enhance cellular differentiation in Candida albicans and Candida tropicalis biofilms. Applied and environmental microbiology 73:4940-4949. Al-Fattani MA, Douglas LJ. 2006. Biofilm matrix of Candida albicans and Candida tropicalis: chemical composition and role in drug resistance. Journal of medical microbiology 55:999-1008. Kothavade RJ, Kura MM, Valand AG, Panthaki MH. 2010. Candida tropicalis: its prevalence, pathogenicity and increasing resistance to fluconazole. Journal of medical microbiology 59:873-880. Douglas LJ. 2003. Candida biofilms and their role in infection. Trends in microbiology 11:30-36. Ramage G, Saville SP, Thomas DP, Lopez-Ribot JL. 2005. Candida biofilms: an update. Eukaryotic cell 4:633-638.

573 574 575 576 577 20

J.Lemire et al. 2015 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626

FIGURE CAPTIONS Figure 1: Silver oxynitrate (Ag7NO11) prevents biofilm formation at lower concentrations versus other metal compounds. The MBEC™ device was inoculated with E.coli (JM109), P.aeruginosa (PAO1), and S.aureus (ATCC 25923) concurrently with serial dilutions (two-fold) of Ag2O, AgO, Ag2SO4, Ag7NO11, AgSD, and CuSO4. The A) MBEC and B) MBC were then determined after a 4h incubation with the various metal compounds. Values are represented as the median ± the range, n=4. * Indicates a significant difference between Ag7NO11 and the indicated metal compound p≤0.05. #Note: all Ag stock solutions were prepared at equal molar equivalents of Ag molecules. Hence, concentrations found in this figure are reflective of the concentration of Ag and not the silver containing compound itself. Figure 2: Silver oxynitrate (Ag7NO11) eradicates established biofilms at lower concentrations versus other metal compounds. The MBEC™ device was inoculated with E.coli (JM109), P.aeruginosa (PAO1), and S.aureus (ATCC 25923). Biofilms were established following a 24h incubation. The established biofilms were then treated with serial dilutions (two-fold) of Ag2O, AgO, Ag2SO4, Ag7NO11, AgSD, and CuSO4 for 24h. The MBEC was then determined for the various metal compounds. Values are represented as the median ± the range, n=4. * Indicates a significant difference between Ag7NO11 and the indicated metal compound p≤0.05. #Note: all Ag stock solutions were prepared at equal molar equivalents of Ag molecules. Hence, concentrations found in this figure are reflective of the concentration of Ag and not the silver containing compound itself. Figure 3: Silver oxynitrate (Ag7NO11) is more efficacious for eradicating mature biofilms of E.coli and S.aureus. E.coli (JM109), P.aeruginosa (PAO1), and S.aureus (ATCC 25923) biofilms were cultivated using the MBEC™ device. Established biofilms were formed following an incubation of 4d for P.aeruginosa, and 6d for E.coli as well as S.aureus. The biofilms were then treated with various concentrations of  CuSO4 (closed line),  AgNO3 (broken line), and Ag7NO11 (dotted line) for 48h. The MBEC was then determined for the various metal compounds. Values are represented as the mean ± the SD, n=3. #Note: all Ag stock solutions were prepared at equal molar equivalents of Ag molecules. Hence, concentrations found in this figure are reflective of the concentration of Ag and not the silver containing compound itself. Figure 4: Silver oxynitrate (Ag7NO11) reduces biofilm quantity. Biofilms of E.coli (JM109), P.aeruginosa (PAO1), and S.aureus (ATCC 25923) were established in the MBEC™ device for 24h. The established biofilms were then exposed to A) 0 μM, B) 5μM and C) 12.5μM AgNO3 or Ag7NO11 (at equivalent Ag concentrations) for 24h. Live/dead™ staining was then performed on the treated biofilms and the biofilm was quantified using a confocal microscope operating at 20X objective magnification. Green (Syto-9; Ex: 488nm, Em: 498nm) = live cells. Red (propidium iodide; Ex:523nm, Em: 617nm) = dead cells. #Note: all Ag stock solutions were prepared at equal molar equivalents of Ag molecules. Hence, concentrations found in this figure are reflective of the concentration of Ag and not the silver containing compound itself. Figure 5: Silver oxynitrate (Ag7NO11) reduces biofilm-biomass. Biofilms of E.coli (JM109), P.aeruginosa (PAO1), and S.aureus (ATCC 25923) were established in the MBEC™ device for 24h. The biofilms were then exposed to various concentrations of A) CuSO4, B) AgNO3 and C) Ag7NO11. Pursuant, a crystal violet staining protocol –using the O’Toole method (53) – was performed on the biofilms. The biomass was quantified by measuring the absorbance at 550 nm. Values are represented as the mean ± the SD, n=15. * Indicates a significant difference between CuSO4 and Ag7NO11, where 21

J.Lemire et al. 2015 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668

* = p≤0.05, ** = p≤0.01, *** is p≤0.001, and **** is p≤0.0001. + Indicates a significant difference between AgNO3 and Ag7NO11, where + = p≤0.05, ++ = p≤0.01, +++ is p≤0.001, and ++++ is p≤0.0001. #Note: all Ag stock solutions were prepared at equal molar equivalents of Ag molecules. Hence, concentrations found in this figure are reflective of the concentration of Ag and not the silver containing compound itself. Figure 6: Silver oxynitrate (Ag7NO11) has antimicrobial and antibiofilm activity against uropathogenic E.coli (UPEC) [CFT703]. UPEC biofilms were cultivated using the MBEC™ device. The A) MBC, on the planktonic population, and B) MBEC, on the biofilm population, were determined after a 4h incubation in the presence of serial dilutions (two-fold) of various metal compounds. C) Established biofilms were cultivated in the MBEC™ device for 24h. The MBEC was then determined following a 24h treatment with serial dilutions (two-fold) of various metal compounds.  CuSO4 (closed line),  AgNO3 (broken line), and Ag7NO11 (dotted line). Values are represented as the mean ± the SD, n=4. #Note: all Ag stock solutions were prepared at equal molar equivalents of Ag molecules. Hence, concentrations found in this figure are reflective of the concentration of Ag and not the silver containing compound itself. . Figure 7: Silver oxynitrate (Ag7NO11) has antimicrobial and antibiofilm activity against a fluoroquinolone-resistant P.aeruginosa (FQRP) isolate. FQRP biofilms were cultivated using the MBEC™ device. The A) MBC, on the planktonic population, and B) MBEC, on the biofilm population, were determined after a 4h incubation in the presence of serial dilutions (two-fold) of various metal compounds. C) Established biofilms were cultivated in the MBEC™ device for 24h. The MBEC was then determined following a 24h treatment with serial dilutions (two-fold) of various metal compounds.  CuSO4 (closed line),  AgNO3 (broken line), and Ag7NO11 (dotted line). Values are represented as the mean ± the SD, n=4. #Note: all Ag stock solutions were prepared at equal molar equivalents of Ag molecules. Hence, concentrations found in this figure are reflective of the concentration of Ag and not the silver containing compound itself. Figure 8: Silver oxynitrate (Ag7NO11) has antimicrobial and antibiofilm activity against a methicillin-resistant S.aureus (MRSA) strain, [USA300]. MRSA biofilms were cultivated using the MBEC™ device. The A) MBC, on the planktonic population, and B) MBEC, on the biofilm population, were determined after a 4h incubation in the presence of serial dilutions (two-fold) of various metal compounds. C) Established biofilms were cultivated in the MBEC™ device for 24h. The MBEC was then determined following a 24h treatment with serial dilutions (two-fold) of various metal compounds.  CuSO4 (closed line),  AgNO3 (broken line), and Ag7NO11 (dotted line). Values are represented as the mean ± the SD, n=4. #Note: all Ag stock solutions were prepared at equal molar equivalents of Ag molecules. Hence, concentrations found in this figure are reflective of the concentration of Ag and not the silver containing compound itself.

22

Silver oxynitrate, an unexplored silver compound with antimicrobial and antibiofilm activity.

Historically it has been accepted, and recent research has established, that silver (Ag) is an efficacious antimicrobial agent. A dwindling pipeline o...
4MB Sizes 0 Downloads 10 Views