Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

You’ve supercharged your research process with ACS and Mendeley!

STEP 1:
Click to create an ACS ID

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

MENDELEY PAIRING EXPIRED
Your Mendeley pairing has expired. Please reconnect
ACS Publications. Most Trusted. Most Cited. Most Read
Potent Antifungal Activity of Penta-O-galloyl-β-d-Glucose against Drug-Resistant Candida albicans, Candida auris, and Other Non-albicans Candida Species
My Activity
CONTENT TYPES

Figure 1Loading Img
  • Open Access
Article

Potent Antifungal Activity of Penta-O-galloyl-β-d-Glucose against Drug-Resistant Candida albicans, Candida auris, and Other Non-albicans Candida Species
Click to copy article linkArticle link copied!

  • Lewis Marquez
    Lewis Marquez
    Molecular and Systems Pharmacology, Laney Graduate School, Emory University, Atlanta, Georgia 30322, United States
    Jones Center at Ichauway, Newton, Georgia 39870, United States
  • Yunjin Lee
    Yunjin Lee
    Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada
    More by Yunjin Lee
  • Dustin Duncan
    Dustin Duncan
    Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada
    Department of Chemistry, Brock University, St. Catharines, Ontario L2S 3A1, Canada
  • Luke Whitesell
    Luke Whitesell
    Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada
  • Leah E. Cowen
    Leah E. Cowen
    Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada
  • Cassandra Quave*
    Cassandra Quave
    Center for the Study of Human Health, Emory University, Atlanta, Georgia 30322, United States
    Department of Dermatology, Emory University, Atlanta, Georgia 30322, United States
    *Email: [email protected]
Open PDFSupporting Information (1)

ACS Infectious Diseases

Cite this: ACS Infect. Dis. 2023, 9, 9, 1685–1694
Click to copy citationCitation copied!
https://doi.org/10.1021/acsinfecdis.3c00113
Published August 22, 2023

Copyright © 2023 The Authors. Published by American Chemical Society. This publication is licensed under

CC-BY 4.0 .

Abstract

Click to copy section linkSection link copied!

Among fungal pathogens, infections by drug-resistant Candida species continue to pose a major challenge to healthcare. This study aimed to evaluate the activity of the bioactive natural product, penta-O-galloyl-β-d-glucose (PGG) against multidrug-resistant (MDR) Candida albicans, MDR Candida auris, and other MDR non-albicans Candida species. Here, we show that PGG has a minimum inhibitory concentration (MIC) of 0.25–8 μg mL–1 (0.265–8.5 μM) against three clinical strains of C. auris and a MIC of 0.25–4 μg mL–1 (0.265–4.25 μM) against a panel of other MDR Candida species. Our cytotoxicity studies found that PGG was well tolerated by human kidney, liver, and epithelial cells with an IC50 > 256 μg mL–1 (>272 μM). We also show that PGG is a high-capacity iron chelator and that deletion of key iron homeostasis genes in C. albicans rendered strains hypersensitive to PGG. In conclusion, PGG displayed potent anti-Candida activity with minimal cytotoxicity for human cells. We also found that the antifungal activity of PGG is mediated through an iron-chelating mechanism, suggesting that the compound could prove useful as a topical treatment for superficial Candida infections.

This publication is licensed under

CC-BY 4.0 .
  • cc licence
  • by licence
Copyright © 2023 The Authors. Published by American Chemical Society

Special Issue

Published as part of the ACS Infectious Diseases virtual special issue “Drug Resistance in Infectious Diseases and Beyond”.

Candida is a genus of commensal yeast that can most often be found in the digestive tract and colonizing the skin of healthy individuals. (1,2) Commensal Candida species can lead to superficial skin candidiasis, with prevalence varying by the geographic region, with reported rates ranging from 22% (China), 40% (Iran), and up to 56% (Serbia). (3−5) When this yeast develops resistance to antifungals and/or invades other parts of the human body, termed invasive candidiasis, serious issues can arise. Invasive candidiasis is often detected as candidemia, a bloodstream infection, but can also involve other major organs such as the heart, kidneys, spleen, and liver. (6) There are an estimated 25,000 infections attributed to Candida each year in the USA. (7) Studies in the USA and Spain have estimated a 29–31% mortality rate for invasive candidiasis. (8,9) Other studies found mortality rates to be twice as high in South Africa (60%) and Brazil (72%). (10,11)
Candida auris, first identified in 2009, is a relatively new Candida species emerging in the landscape of invasive candidiasis. C. auris infections are highly invasive and deadly, (12,13) with mortality rates as high as 60%. (14,15) Most recently, from June 2020 to May 2021, there were more than 1,000 clinical cases of C. auris reported across 19 states in the USA. (16)
Drug resistance and a paucity of new antifungals in development necessitate the discovery of new anti-Candida drugs. Preparations from Schinus terebinthifolia Raddi (Anacardiaceae family), the Brazilian peppertree, have traditionally been used as an antiseptic and for the treatment of skin ulcers and wound healing. (17) The present study investigated a bioactive small molecule with anti-Candida activity that our lab first isolated from the leaves of S. terebinthifolia─the ubiquitous natural product penta-O-galloyl-β-d-glucose (PGG). (18) PGG is a hydrolyzable tannin with a plethora of biological activities such as antibacterial, anticancer, and antiviral activities (Table 1). We examined PGG for cytotoxicity against a panel of human cell lines and growth inhibition against multidrug-resistant (MDR) strains of Candida albicans, Candida glabrata, Candida parapsilosis, and C. auris (Table 2). Additionally, we sought to elucidate the mechanism of action for the antifungal activity of PGG.
Table 1. Notable Biological Activities of Penta-O-galloyl-β-d-glucose (PGG)
targetIC50/EC50mode of actionrefs
Antibacterial
Staphylococcus aureus (various strains)16–32 μg/mLpossibly iron chelation (19)
MRSA (various strains)16–32 μg/mLpossibly iron chelation (19)
Acinetobacter baumannii (various strains)16–64 μg/mLpossibly iron chelation (18)
Anticancer (Liver Cancer)
SK-HEP-1 cells25–50 μMinhibition of proliferation (inactivation of NF-κB) (20)
Anticancer (Colorectal Cancer)
HCT-116 cells1.61 μMinhibition of proliferation (induction of the p53 gene) (21,22)
HT-29 cells4.46 μMinhibition of proliferation (induction of the p53 gene) (21,22)
Anticancer (Breast Cancer)
MDA-MB-23150.23 μMinduction of apoptosis (23)
MDA-MB-46835.72 μMinduction of apoptosis (23)
Antiviral
influenza A5.3 μg/mLinterference with viral replication (24)
Table 2. MIC Values for PGG against the Various Candida Species and Strains
   IC50 (μg/mL)MIC (μg/mL)MIC (μg/mL)
speciesstrain IDantibiogramPGGPGGAMBFLCKTC
Candida albicansATCC 90028AfgS, CasS, FlcS, ItcS, MfgS, VrcS0.5110.25<0.0039
Candida albicansMH12AfgS, CasS, FlcR, ItcS, MfgS, VrcS0.250.51>81
Candida parapsilosisAR Bank #0335AfgI, CasS, FlcR, MfgS, VrcR141>84
Candida parapsilosisAR Bank #0337AfgS, CasS, FlcR, MfgS, VrcR0.50.51>82
Candida parapsilosisAR Bank #0338AfgS, CasS, FlcR, MfgS0.250.51>80.5
Candida parapsilosisAR Bank #0339AfgS, CasS, FlcR, MfgS0.512>80.25
Candida glabrataAR Bank #0325AfgR, CasR, FlcR, MfgR0.250.51>84
Candida glabrataAR Bank #0326AfgS, CasS, FlcSDD, MfgS,0.1250.25140.25
Candida aurisaAR Bank #0381AfgS, AmbS, CasS, FlcS, MfgS0.2514<0.0039
Candida aurisaAR Bank #0387AfgS, AmbS, CasS, FlcI, MfgS0.510.51<0.0039
Candida aurisaAR Bank #0390AfgS, AmbR, CasS, FlcR, MfgS482>80.5
a

There are currently no established C. auris antifungal breakpoints; as outlined by the CDC, (25) breakpoints from closely related Candida sp. were used to determine the antibiogram profile. Abbreviations used are amphotericin B (Amb), anidulafungin (Afg), caspofungin (Cas), itraconazole (Itc), fluconazole (Flu), micafungin (Mfg), voriconazole (Vrc), and ketoconazole (Ktc). Superscripts used: S, susceptible; SDD, susceptible dose-dependent; I, intermediate; and R, resistant.

Results

Click to copy section linkSection link copied!

Anti-Candida Activity of Penta-O-galloyl-β-d-glucose

Penta-O-galloyl-β-d-glucose (PGG, Figure 1) was tested for growth inhibition using Clinical and Laboratory Standards Institute (CLSI) guidelines against seven drug-resistant strains and four susceptible strains from four species of Candida: two strains of C. albicans, two strains of C. glabrata, four strains of C. parapsilosis, and three strains of C. auris (Table 2). We found that MICs against all Candida species tested ranged from 0.25–8 μg mL–1 (equivalent to 0.265–8.5 μM) (Table 2). PGG MICs for C. albicans ranged from 0.5 to 1.0 μg mL–1 (Figure 2A), for C. glabrata ranged from 0.25 to 0.5 μg mL–1 (Figure 2B), for C. parapsilosis ranged from 0.5 to 4.0 μg mL–1 (Figure 2C), and for C. auris ranged from 0.25 to 8.0 μg mL–1 (Figure 2D).

Figure 1

Figure 1. Chemical structure of penta-O-galloyl-β-d-glucose, PGG.

Figure 2

Figure 2. Penta-O-galloyl-β-d-glucose displays broad spectrum anti-Candida activity. Growth inhibition of PGG against (A) C. albicans, (B) C. glabrata, (C) C. parapsilosis, and (D) C. auris. Solid lines represent PGG. Dashed lines represent the positive control fluconazole. The data are plotted as the mean % growth inhibition ± SD as compared to the DMSO vehicle from two independent experiments. The horizontal dotted line represents 90% growth inhibition. See Supplemental Figures S8–S11 for expanded graphs including additional positive controls. See Table 2 for strain details.

We also examined PGG for the synergistic interaction with two common antifungals and found that PGG does not potentiate the activity of the echinocandin caspofungin or the azole fluconazole against C. albicans (Supplemental Figure S1).

Cytotoxicity

We examined PGG’s effects on cell viability and cytotoxicity against three human cell lines: human embryonic kidney 293 cells (Hek293), human hepatoblastoma cells (HepG2), and human epidermal keratinocytes (HaCaT). In cytotoxicity studies with Hek293, HepG2, and HaCaT cells, we found that PGG was well tolerated with a CC50 > 256 μg mL–1 (>272 μM) (Supplemental Figure S2). We assessed relative viable cell number by measuring relative ATP levels in vitro after treatment with PGG and found that PGG reduced the viable cell number by 50% in Hek293 and HepG2 cells at an IC50 of 64 μg mL–1 (68 μM) and displayed an IC50 of 32 μg mL–1 (34 μM) against HaCaT cells (Supplemental Figure S3).

PGG Sensitivity of C. albicans Is Not Modulated by Major Efflux Pumps

To determine if PGG activity was affected by efflux mediated by four of the major C. albicans efflux pumps, we tested PGG against a mutant strain of C. albicans with deletion of the genes encoding these efflux pumps: Candida drug-resistance genes (CDR1, CDR2), fluconazole resistance gene (FLU1), and (MDR1) multidrug resistance gene. (26) We found that the absence of these major efflux pumps did not modify sensitivity to PGG, in contrast to the hypersensitivity seen with fluconazole, a compound known to undergo efflux pump-mediated transport (Figure 3). These data provide evidence that PGG is not a substrate for these major C. albicans efflux pumps and/or does not need to enter the cell to produce its effect.

Figure 3

Figure 3. PGG sensitivity in C. albicans is not modulated by efflux pumps. The cdr1Δ/Δ, cdr2Δ/Δ, mdr1Δ/Δ, flu1Δ/Δ, and wild-type strains were grown overnight in YPD and subject to dose–response assays in RPMI-1640 medium with twofold dilution gradients of PGG. Fluconazole was included as a positive control. Growth (OD600nm) was measured after 72 h of incubation at 30 °C, averaged between technical duplicates, and normalized to the drug-free wild-type control. Results are presented in a heat-map format with a scale bar provided at the bottom of the figure. All assays were performed in biological duplicates.

PGG Does Not Bind to Fungal Ergosterol

Probing for possible mechanisms of action, we examined the interaction of PGG with ergosterol, a sterol found in the fungal cell membrane and the major target of the polyene class of antifungals. We found that PGG’s growth inhibitory activity was not affected by additional ergosterol, providing evidence that PGG does not target ergosterol in the fungal cell membrane (Supplemental Figure S4).

PGG’s Anti-Candida Activity Is Tied to Iron Chelation

A previous study by Cho et al. (2010) found a possible link between the antibacterial activity of PGG and iron chelation in cultures of Staphylococcus aureus. (19) To determine if iron chelation was also relevant to the antifungal activity of PGG against Candida, we tested C. albicans, C. glabrata, and C. auris grown in iron-supplemented media. We found that the inhibitory effect of PGG against C. albicans, C. glabrata, and C. auris was abrogated upon the addition of iron supplementation (Figure 4A–C). C. albicans, C. glabrata, and C. auris supplemented with 10 mM of iron (II) or iron (III) led to a reduction of approximately 80–100% of the growth inhibitory activity of PGG at concentrations of 4–16 μg mL–1. We next examined the specificity of PGG for other metals and cations such as manganese, zinc, calcium, and magnesium. We found no significant effect on growth inhibition from cation treatment, no effect from zinc supplementation, and a minor effect on growth inhibition from manganese supplementation at 0.5× MIC of PGG (Supplemental Figures S5, S6). These results provide evidence that PGG’s activity is selective for iron.

Figure 4

Figure 4. Growth inhibition of PGG against multiple Candida species is abrogated by iron supplementation, and PGG binds multiple iron ions. (A) C. albicans 90028, (B) C. glabrata CDC325, and (C) C. auris CDC381 were treated with PGG or Amphotericin B at varying concentrations (4, 8, and 16 μg/mL) and media was supplemented with a 10 mM solution of ferrous sulfate (Fe2+) or ferric sulfate (Fe3+) and % growth inhibition measured after 48 h. The data are plotted as the mean % growth inhibition ± SD as compared to the DMSO vehicle from two independent experiments. The horizontal dotted line represents 90% growth inhibition compared to the vehicle. Statistical analysis was done using two-way ANOVA with multiple comparisons. (D) PGG or EGCG was allowed to react with varying concentrations of iron (III) in solution at pH 2.0. The bipy solution was added to the mixtures and the formation of the [Fe(bipy)3]2+ complex was measured at 520 nm. [PGG]total, [EGCG]total = 10 μM. The data are plotted as the mean absorbance ± SD from two independent experiments.

To further investigate the role of iron chelation as it relates to PGG’s anti-Candida activity, we next examined the iron-binding capacity of PGG. We utilized the ligand 2,2′-bipyridyl (bipy). The bipy method utilized a proxy of PGG-iron binding through the spectroscopic determination of bipy–iron complex formation. Bipy is a well-known ligand that can measure, from mixtures of iron (III) and a reducing agent, the amount of free iron (II) as a red-colored complex [Fe(bipy)3]2+ with absorbance at 520 nm. From this experiment, we can calculate the amount of iron (III) ions bound by the galloyl moieties on the PGG molecule through the reduction of iron (III) to iron (II) and the detection of the colored [Fe(bipy)3]2+ complex in a 1:1 ratio. Results of the 2,2′-bipyridyl test found that in the presence of excess iron (III), PGG bound more than 2× the amount of iron (III) compared to epigallocatechin-gallate (EGCG), as shown in Figure 4D. EGCG is a natural product with two galloyl side groups, as compared to the five galloyl side groups on PGG. EGCG has previously been shown to bind up to two iron (III) ions per molecule of EGCG. (27) Calculations of these results equate to the reduction of 10 Fe3+ ions to 10 Fe2+ ions and the binding of 5 iron (III) ions per PGG molecule at the highest tested concentrations. We also attempted to determine stoichiometry using Job’s method. (28,29) Interestingly, we found that depending on the mole fraction of iron and PGG, the peak wavelength varied (Supplemental Figure S7A). This suggests that at different mole fractions of iron and PGG, different complexes form. Since Job’s method assumes that only one complex is formed, we were unable to determine a discrete stoichiometry of PGG to iron but were still able to demonstrate that PGG was able to bind multiple iron ions (Supplemental Figure S7B). These results provide evidence for the high iron-binding capacity of each PGG molecule and the ability to form multiple complexes with iron.
Given that iron rescues PGG growth inhibition, we next examined whether key iron homeostasis genes in C. albicans are required for basal tolerance to PGG. We tested PGG’s effect against multiple iron homeostasis gene mutants and found that deletion of IRE1, which encodes a protein kinase with an essential function in iron uptake, as well as HAP43 and RIM101, which encode transcription factors involved in the iron-starvation response, (30−32) rendered strains hypersensitive to PGG, as is the case with a known iron chelator, bathophenanthrolinedisulfonate (BPS) (Figure 5). Together, these results support the conclusion that PGG’s antifungal activity is linked to iron chelation.

Figure 5

Figure 5. Iron homeostasis genes are required for PGG tolerance in C. albicans. The ire1Δ/ire1Δ, hap43Δ/hap43Δ, and rim101Δ/rim101Δ strains and their respective wild-type controls were grown overnight in YPD and subject to dose–response assays in RPMI-1640 medium with twofold dilution gradients of PGG. The metal chelator BPS was included as a positive control. After 48 h incubation at 30 °C, the relative viable cell number was assessed. Data were averaged between technical duplicates and normalized to wild-type drug-free control wells. Results are presented in a heat-map format with a color scale bar presented at the bottom of the figure. All assays were performed in biological duplicates.

Discussion

Click to copy section linkSection link copied!

Penta-O-galloyl-β-d-glucose (PGG) is a hydrolyzable tannin with many functional roles, such as antibacterial, anticancer, and antiviral activities (Table 1). This study is the first to investigate PGG’s antifungal activity against a panel of multiple Candida species. Our findings show that PGG displays potent activity against C. auris, C. albicans, C. glabrata, and C. parapsilosis with MICs in the range of 0.25–8 μg mL–1 (0.265–8.5 μM). Our findings show that PGG is comparable to, or more effective than, conventional antifungals against multiple drug-resistant strains of Candida species when tested under standard microbiological assay conditions.
We found that PGG displayed a low level of in vitro cytotoxicity (CC50 > 256 μg mL–1) against human kidney, liver, and skin cells in a serum-supplemented tissue culture medium. For a majority of Candida species and strains tested, PGG displayed a MIC between 0.25–1 μg mL–1 (0.265–1.06 μM). However, after further testing of PGG against drug-sensitive C. albicans in fungal growth media supplemented with 10% fetal bovine serum (FBS), we saw evidence of serum-induced MIC reversal and the loss of growth inhibition of PGG at concentrations up to 512 μg mL–1 (data not shown). Of note, the presence of FBS in our human cell cytotoxicity studies could have influenced the results of our cytotoxicity assays through iron supplementation or protein binding of PGG, potentially masking any cytotoxic effects. Despite these concerns, while we did not investigate the in vivo tolerability of PGG, another study found PGG to be tolerated without ill effects at doses of 20 mg/kg/day orally in mice, and a separate study found a dose up to 120 mg/kg/day intravenously in rats produced no ill effects. (33,34) Most recently, PGG was used in humans to safely treat abdominal aortic aneurysms via a balloon catheter at a rate of 3 mg/mL over 25 mL. (35) Our findings, coupled with the aforementioned in vivo studies, suggest that PGG is a compound that is well tolerated both in vitro and in vivo.
One major weakness of PGG is the high clearance rate and breakdown of the compound in vivo. A separate study found that PGG exhibited a high efflux ratio, which provides evidence that PGG likely undergoes active transport out of the cell. (36) Another study found that PGG could not be detected in the plasma after oral administration in mice. (37) The breakdown of PGG into various gallic acid derivatives and metabolites in vivo may be the reason PGG was below the level of detection in plasma and may contribute to its high clearance rate in vivo. (38) For these reasons, PGG, in its current form, likely makes a poor candidate as a drug for systemic administration. However, in line with the traditional ethnobotanical topical use of the leaves of S. terebinthifolia, the broad anti-Candida activity of PGG merits further examination in vivo to evaluate any potential as a topical antifungal agent.
The mechanism of action for the antimicrobial effect of PGG is likely attributable to its activity as an iron chelator. We expect that iron binding occurs via the hydroxyl groups on the galloyl moieties, a trait shared with epigallocatechin-gallate (27) and other similarly structured compounds. (39) Iron is essential for most living organisms, as it is a key component in a variety of metalloenzymes for processes such as cell growth, electron transport, and DNA synthesis. Iron is also essential for the growth and survival of many fungi, including Candida. (40−42) Uptake of extracellular iron, through whatever means, is a crucial process necessary for the growth and survival of Candida species. We observed that excess free iron rescues PGG growth inhibition and genetic deletion of key iron homeostasis genes such as IRE1, HAP43, and RIM101 in C. albicans conferred hypersensitivity to PGG. Our iron-binding experiment found that PGG strongly interacts with free iron, with each molecule of PGG binding up to five iron (III) ions. PGG’s iron-chelating ability is expected to be the mechanism through which PGG elicits its growth inhibitory effect against the tested Candida species. Preventing Candida species from acquiring this crucial micronutrient is an effective means of preventing their proliferation.
There is one other example of an iron-chelating antifungal, the 50-year-old topical antifungal drug ciclopirox. (43,44) Despite the extensive use of ciclopirox as a topical antifungal for over 50 years, there have been no reported ciclopirox-resistant strains in the literature. While the mechanism of action for ciclopirox was initially unknown, there has been ample evidence pointing to its antifungal activity being mediated through an iron-chelating mechanism. (45−47) Sigle et al. (47) found that the antifungal activity of ciclopirox was abrogated upon the addition of both Fe2+ and Fe3+, similar to what we found to occur for our compound PGG. Importantly, PGG is superior to ciclopirox in its iron-binding capacity─three molecules of ciclopirox bind one iron ion, whereas we found one molecule of PGG binds up to five. (48) Sigle et al. (47) found multiple iron metabolism genes upregulated in response to ciclopirox treatment. One of these genes was the unknown transcription factor IPF7711, now known as HAP43. (49) HAP43 (alias IPF7711) is involved in the iron-starvation response (31) and is one of the genes for which we found that deletion confers hypersensitivity to PGG in C. albicans. Regarding the development of resistance to an iron-chelating antifungal agent, Niewerth et al. (45) found that ciclopirox resistance did not develop in C. albicans over 6 months of treatment in vitro. This is similar to the trend we found previously, in which the gram-negative bacteria Acinetobacter baumanii did not develop resistance to PGG over 21 daily passages. (18) The continued clinical use of the iron-chelating topical antifungal ciclopirox, without the emergence of resistant strains, provides logical evidence that the iron-chelating compound PGG could also be developed into an effective topical antifungal agent.
As a final note, our review of the literature on the other biological activities of PGG (Table 1) leads us to suggest that iron chelation may also play a role in PGG’s anticancer activity. The authors Oh et al. (20) found that PGG inhibited the activation of nuclear factor kappa B (NF-κB), a transcription factor involved in the regulation of inflammation and cell proliferation. (50) We theorize that the inactivation of NF-κB by PGG may be due to the iron-chelating activity of PGG. It has been reported that intracellular iron is involved in the activation of NF-κB, (51) and a separate study found that iron activates NF-κB in Kupffer cells. (52) The downstream inactivation of NF-κB and the evident inhibition of proliferation in SK-HEP-1 hepatoma cells may be due to the chelating activity of PGG sequestering iron and modulation of upstream regulators of NF-κB. Other studies examining colorectal cancer cells found that PGG inhibits the proliferation of HCT-116 and HT-29 cells. (21) PGG’s anticancer effect against colorectal cancer cells was proposed to be acting through induction of the tumor suppressor protein p53. (22) Iron metabolism is reported to regulate the activation of p53. Studies have reported that iron depletion by iron chelators promotes activation of p53. (53,54) It is possible, for some cancer types, that the anticancer activity of PGG may initially stem from iron chelation and the anticancer activity reported may be a downstream effect of iron depletion. PGG’s iron-chelating ability may be the basis for many of the biological roles reported for PGG, but more studies are needed to support this connection.
PGG’s strong anti-Candida activity and its demonstrated iron-chelating activity warrant further investigation as a topical therapeutic agent for use against Candida infections. A topical PGG formulation could be beneficial in the treatment of other Candida skin infections or fungal infections found in warm, moist creased skin environments such as the armpits and groin. Studies have shown relatively high recovery yields of PGG (36%) from Mangifera indica L. kernels and mango seeds, an unused food element that could greatly reduce acquisition costs. (55) For these reasons, PGG’s properties as an anti-Candida agent warrant a deeper investigation into its potential therapeutic applications.

Methods

Click to copy section linkSection link copied!

Cell Lines and Growth Conditions

Fungal strains tested were purchased from the American Type Culture Collection (ATCC) or acquired from the FDA-CDC Antimicrobial Resistance Isolate Bank. A full list of tested strains and their antimicrobial susceptibility profiles can be found in Table 2. Strains were maintained on BBL Sabouraud dextrose agar (SDA, BD) at 35 °C in a humidified chamber for 48 h before use. The C. albicans DSY1024 (cdr/cdrcdr/cdrmdr/mdrflu/flu1Δ) mutant, (26) C. albicans ire1Δ/Δ mutant, (56) and C. albicans hap43Δ/Δ and C. albicans rim101Δ/Δ mutants (57) were obtained from previous studies. For all other assays, strains are listed in Supplemental Table S1, along with their sources. Strains were cultured in YPD (1% yeast extract, 2% peptone, and 2% glucose) at 30 °C.
Hek293 cells were purchased from BEI Resources. HaCaT cells were purchased from ATCC. HepG2 cells were provided by Dr. Edward Morgan (Emory University). Mammalian cell cultures were maintained in Dulbecco’s modified Eagle’s medium with L-glutamine and 4.5 g L–1 glucose (Corning) supplemented with 10% fetal bovine serum (Avantor) and a 100× combination solution of 10,000 IU mL–1 penicillin and 10,000 μg mL–1 streptomycin (Corning) at 37 °C, 5% CO2. Mammalian cultures at 70–80% confluency were used for cell viability and cytotoxicity experiments.

Growth Inhibition

Growth inhibition assays were performed by microbroth dilution following the guidelines set by the CLSI. (58) Briefly, Candida spp. was maintained on BBL Sabouraud dextrose agar (SDA) at 35 °C for 48 h. Inoculums were standardized to 2 × 103 CFU mL–1 in RPMI-1640 medium. Wells were treated with penta-O-galloyl-β-d-glucose (PGG, 0.0039–8.0 μg mL–1) (≥96% purity, Sigma Aldrich), positive controls (0.0039–8.0 μg mL–1), or an equivalent volume of the DMSO vehicle. Percent growth inhibition was determined by comparing treatments to the DMSO vehicle control. MIC was determined by measuring OD600 with a BioTek Cytation3 plate reader after incubation (35 °C) with treatment for 48 h. MIC was defined as the lowest treatment concentration with >90% growth inhibition. The IC50 was determined by the lowest treatment concentration with >50% growth inhibition. Positive controls were fluconazole (MP Biomedicals), ketoconazole, and amphotericin B (Alfa Aesar). The negative control was untreated culture. A media blank was used to monitor for media contamination. Tests were repeated in duplicate with three technical replicates per treatment and on different days.
For media supplementation experiments, magnesium sulfate (Alfa Aesar), manganese sulfate, zinc sulfate, and calcium chloride (Ward Science) were dissolved in RPMI-1640 media and, after 24 h, added at a final concentration of 1 mM. Growth inhibition was measured 24 h after media supplementation. Percent growth inhibition was determined by comparing treatments to vehicle controls. Media supplementation experiments were repeated in triplicate with four technical replicates per treatment and on different days.
For iron supplementation assays, growth inhibition assays were performed by microbroth dilution following the guidelines set by the CLSI. (58) A solution of iron (II) sulfate and iron (III) sulfate (VWR) was dissolved in deionized water to a concentration of 10 mM and added to treated wells at the beginning of incubation. Growth inhibition was measured after 48 h. Percent growth inhibition was determined by comparing treatments to vehicle controls. Iron supplementation experiments were repeated in duplicate with three technical replicates per treatment and on different days.

Method for Concentration-Response Matrixes (Checkerboard Assays)

Cells were grown overnight in yeast extract peptone dextrose (YPD) at 30 °C under shaking conditions (200 rpm). Checkerboard assays were set up in 96-well flat-bottom microtiter plates (Sarstedt) in a total well volume of 0.1 mL. Cell inoculums were prepared from saturated overnight cultures at 2 × 104 CFU mL–1, which is 10-fold higher than the inoculums used in the CLSI MICs to increase the dynamic range of the assay. Twofold serial dilutions of compounds in RPMI–1640 medium were prepared along the y- and x-axes. Plates were incubated at 30 °C in static conditions, and growth was assessed by measuring absorbance (OD600) after 48 h. Growth was corrected for the medium background and normalized to the no-drug controls. All assays were performed in technical duplicates in two biological replicates. Data was quantitatively displayed using Java Treeview3. The fractional inhibitory concentration index (FICI) at 90% growth inhibition was calculated to evaluate the interaction of the two drugs in combination, with values <0.5 indicating a synergistic interaction. (59)

Method for the Ergosterol Binding Assay

Ergosterol binding was measured using a previously described method, (60) with some modifications. Briefly, ergosterol (TCI) was pulverized with a sterile pestle and mortar and then dissolved in 100% ethanol. The resultant emulsion was warmed at 37 °C for 15 min and then sonicated for 45 min prior to use. PGG (Sigma Aldrich) and the positive control amphotericin B (Alfa Aesar) were serially diluted to a final concentration equal to 1× – 4× the MIC of PGG (1.0–4.0 μg mL–1). Growth inhibition was determined in the presence and absence of 80 μg mL–1 of exogenous ergosterol by broth microdilution following the guidelines set by the CLSI. (58) Percent growth inhibition was measured after 24 h of incubation by comparing treatments to vehicle controls. The negative control was untreated culture. A media blank was used to monitor for contamination. Experiments were repeated in triplicate with three technical replicates per treatment and on different days.

Concentration-Response Assays of C. albicans Efflux and Iron Homeostasis Mutants

Strains were grown overnight in YPD at 30 °C under shaking conditions (200 rpm). Cell inoculums were prepared in RPMI-1640 medium at 2 × 104 CFU mL–1, which is 10-fold higher than the inoculums used in the CLSI MICs to increase the dynamic range of the assay. Concentration-response assays were performed in RPMI-1640 medium using a broth microdilution protocol, as previously described. (61) Plates were incubated at 30 °C in static conditions for 48 or 72 h, as indicated. For the efflux mutants, growth was measured by absorbance (OD600). For the iron homeostasis mutants, growth was measured by Alamar blue (Invitrogen) fluorescence at 530/590 nm (excitation/emission) to circumvent issues with optical density quantification of filamentous cells. Growth was corrected for the medium background and normalized to the no-drug wild-type controls. All assays were performed in technical duplicates in two biological replicates. Data was quantitatively displayed using Java Treeview3.

2,2′-Bipyridyl Test

The 2,2′-bipyridyl test was followed and analyzed as outlined previously (27) and modified for a 96-well plate format. Briefly, 2,2′-bipyridyl (Sigma Aldrich) interacts with free iron (II) to produce a red-colored complex [Fe(bipy)3]2+ with absorbance at 520 nm. A 0:1 solution of iron (III):ligand was prepared at starting concentrations of 10 μM, adjusted to 2.0 pH, and allowed to equilibrate at room temperature for 1 h. This was repeated for 1:1, 2:1, 3:1, etc. iron (III):ligand mixtures. A solution of 2,2′-bipyridyl was added to the iron (III):ligand mixture and mixed at 650 rpm for 5 min. Absorbance was read with a Cytation3 plate reader with path length correction selected. Solutions of ligand only were used as a blank. The positive control was epigallocatechin-gallate (EGCG, Cayman Chemical). The negative control was a solution of only 2,2′-bipyridyl. The molar absorptivity of [Fe(bipy)3]2+ was previously found to be 8200 M–1 cm–1. (27) The number of Fe2+ ions produced and the number of electron transfers that occurred were then calculated with the following equation:
NumberofFe2+ionsproduced=Aεbc
where A = absorbance, ε = molar absorptivity of [Fe(bipy)3]2+ (8200 M–1 cm–1), b = path length of the beam (1 cm), and c = concentration of the ligand (0.00001 M).

Method for Job’s Method

In a 384-well plate, the ligand and metal were added to a 1:1 DMSO:H2O solution in varying mole ratios to a total ligand and metal concentration of 0.76 mM (PGG) from 7.6 mM stock concentrations to a total volume of 100 μL. The mixtures were incubated at room temperature for approximately 5 min, and each well was scanned between 400 and 750 nm at 2 nm increments.

Cell Viability and Cytotoxicity

Chemical-Induced Cytotoxicity

The mammalian culture was standardized to 4 × 103 cells per 100 μL. Cells were allowed to attach and incubate for 24 h before growth media was replaced with treatment media (PGG, 16–512 μg mL–1) or an equivalent volume of the DMSO vehicle, followed by incubation for 24 h. Plates were then evaluated with an LDH cytotoxicity assay (G Biosciences) and processed according to the manufacturer’s protocol for chemical-induced cytotoxicity. Experiments were repeated in triplicate with three technical replicates per treatment and on different days.

Cell Viability/Proliferation

ATP content as a marker for the relative viable cell number was measured with the CellTiter-Glo Luminescent Cell Viability assay (Promega Corporation). The mammalian culture was standardized at 4 × 103 cells per 100 μL in a Costar 3610 white plate, clear bottom 96-well plates (Corning). Cells were allowed to attach and incubate for 24 h before growth media was replaced with treatment media (PGG, 16–512 μg mL–1) or an equivalent volume of DMSO, followed by incubation for 24 h. After incubation with treatment, plates were equilibrated to room temperature for 1 h, and then 100 μL of CellTiter-Glo reagent was added to each well and mixed for 2 min. Plates were allowed to incubate at room temperature for 10 min before luminosity was measured with an integration time of 1 s via a BioTek Cytation3 plate reader. The relative viable cell number was measured by comparing treatment samples to untreated controls. Experiments were repeated in triplicate with three technical replicates per treatment and on different days.

Statistical Analysis

Where applicable, data were analyzed by two-way ANOVA with Tukey’s multiple comparison test using GraphPad Prism version 9.30 for Windows, GraphPad Software, San Diego, California, USA, www.graphpad.com. A p-value <0.05 was considered significant.

Data Availability

Click to copy section linkSection link copied!

ChemDraw Professional version 16.0.1.4 (77) was used for chemical structures.

Supporting Information

Click to copy section linkSection link copied!

The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsinfecdis.3c00113.

  • Experimental methods for concentration-response matrixes; effects of penta-O-galloyl-β-d-glucose upon human cytotoxicity and cell viability; media supplementation assays; Job’s method spectra; expanded MIC graphs with additional positive controls; and table with the additional Candida albicans strains and genotype (PDF)

Terms & Conditions

Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

Author Information

Click to copy section linkSection link copied!

  • Corresponding Author
    • Cassandra Quave - Center for the Study of Human Health, Emory University, Atlanta, Georgia 30322, United StatesDepartment of Dermatology, Emory University, Atlanta, Georgia 30322, United States Email: [email protected]
  • Authors
    • Lewis Marquez - Molecular and Systems Pharmacology, Laney Graduate School, Emory University, Atlanta, Georgia 30322, United StatesJones Center at Ichauway, Newton, Georgia 39870, United StatesOrcidhttps://orcid.org/0000-0002-3782-5204
    • Yunjin Lee - Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada
    • Dustin Duncan - Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, CanadaDepartment of Chemistry, Brock University, St. Catharines, Ontario L2S 3A1, CanadaPresent Address: Department of Chemistry, Brock UniversityOrcidhttps://orcid.org/0000-0003-1240-8495
    • Luke Whitesell - Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada
    • Leah E. Cowen - Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, CanadaOrcidhttps://orcid.org/0000-0001-5797-0110
  • Author Contributions

    L.M., L.W., L.E.C., and C.L.Q. conceived and designed the experiments, L.M. performed MIC assays of Candida species, cell cytotoxicity/viability assays, media supplementation assays, ergosterol assay, 2,2′-bipyridyl test, and data analysis, Y.L. performed the concentration-response matrixes (checkerboard assays) and concentration-response assays of C. albicans efflux and iron homeostasis mutants, D.D performed the Job’s method experiments, L.M. prepared the first draft, and all authors reviewed the results, edited the draft, and approved the final version of the manuscript.

  • Notes
    The authors declare the following competing financial interest(s): C.L.Q. and L.M. are inventors on a patent pertaining to the use of PGG for the mitigation of fungal infections. L.E.C. and L.W. are a co-founders and shareholders in Bright Angel Therapeutics, a platform company for development of antifungal therapeutics. L.E.C. is a Science Advisor for Kapoose Creek, a company that harnesses the therapeutic potential of fungi.

Acknowledgments

Click to copy section linkSection link copied!

This work was supported in part by a grant from the National Institutes of Health, National Center for Complementary and Integrative Health (NCCIH) (R01AT011990 to C.L.Q.) and a graduate student fellowship from The Jones Center at Ichauway to L.M. Y.L. is supported by the CIHR Frederick Banting and Charles Best Canada Graduate Scholarship─Doctoral Awards. L.E.C. is supported by the Canadian Institutes of Health Research Foundation Grant (FDN-154288) and is a Canada Research Chair (Tier 1) in Microbial Genomics & Infectious Disease and co-director of the CIFAR Fungal Kingdom: Threats & Opportunities program.

References

Click to copy section linkSection link copied!

This article references 61 other publications.

  1. 1
    Limon, J. J.; Skalski, J. H.; Underhill, D. M. Commensal Fungi in Health and Disease. Cell Host Microbe 2017, 22, 156165,  DOI: 10.1016/j.chom.2017.07.002
  2. 2
    Neville, B. A.; d’Enfert, C.; Bougnoux, M.-E. Candida albicans commensalism in the gastrointestinal tract. FEMS Yeast Res 2015, 15, fov081,  DOI: 10.1093/femsyr/fov081
  3. 3
    Bilal, H.; Hou, B.; Shafiq, M.; Chen, X.; Shahid, M. A.; Zeng, Y. Antifungal susceptibility pattern of Candida isolated from cutaneous candidiasis patients in eastern Guangdong region: A retrospective study of the past 10 years. Front. Microbiol. 2022, 13, 981181  DOI: 10.3389/fmicb.2022.981181
  4. 4
    Khodadadi, H.; Zomorodian, K.; Nouraei, H.; Zareshahrabadi, Z.; Barzegar, S.; Zare, M. R.; Pakshir, K. Prevalence of superficial-cutaneous fungal infections in Shiraz, Iran: A five-year retrospective study (2015–2019). J. Clin. Lab. Anal. 2021, 35, e23850  DOI: 10.1002/jcla.23850
  5. 5
    Otašević, S.; Momčilović, S.; Golubović, M.; Ignjatović, A.; Rančić, N.; Đorđević, M.; Ranđelović, M.; Hay, R.; Arsić-Arsenijević, V. Species distribution and epidemiological characteristics of superficial fungal infections in Southeastern Serbia. Mycoses 2019, 62, 458465,  DOI: 10.1111/myc.12900
  6. 6
    Pappas, P. G.; Lionakis, M. S.; Arendrup, M. C.; Ostrosky-Zeichner, L.; Kullberg, B. J. Invasive candidiasis. Nat. Rev. Dis. Primers 2018, 4, 18026,  DOI: 10.1038/nrdp.2018.26
  7. 7
    Tsay, S.; Williams, S.; Mu, Y.; Epson, E.; Johnston, H.; Farley, M. M.; Harrison, L. H.; Vonbank, B.; Shrum, S.; Dumyati, G.; Zhang, A.; Schaffner, W.; Magill, S.; Vallabhaneni, S. 363. National Burden of Candidemia, United States, 2017. Open Forum Infect. Dis. 2018, 5, S142S143,  DOI: 10.1093/ofid/ofy210.374
  8. 8
    Puig-Asensio, M.; Padilla, B.; Garnacho-Montero, J.; Zaragoza, O.; Aguado, J. M.; Zaragoza, R.; Montejo, M.; Muñoz, P.; Ruiz-Camps, I.; Cuenca-Estrella, M.; Almirante, B. Epidemiology and predictive factors for early and late mortality in Candida bloodstream infections: a population-based surveillance in Spain. Clin. Microbiol. Infect. 2014, 20, O24554,  DOI: 10.1111/1469-0691.12380
  9. 9
    Cleveland, A. A.; Farley, M. M.; Harrison, L. H.; Stein, B.; Hollick, R.; Lockhart, S. R.; Magill, S. S.; Derado, G.; Park, B. J.; Chiller, T. M. Changes in incidence and antifungal drug resistance in candidemia: results from population-based laboratory surveillance in Atlanta and Baltimore, 2008-2011. Clin. Infect. Dis. 2012, 55, 13521361,  DOI: 10.1093/cid/cis697
  10. 10
    Kreusch, A.; Karstaedt, A. S. Candidemia among adults in Soweto, South Africa, 1990-2007. Int. J. Infect. Dis. 2013, 17, e6213,  DOI: 10.1016/j.ijid.2013.02.010
  11. 11
    Doi, A. M.; Pignatari, A. C. C.; Edmond, M. B.; Marra, A. R.; Camargo, L. F. A.; Siqueira, R. A.; da Mota, V. P.; Colombo, A. L. Epidemiology and Microbiologic Characterization of Nosocomial Candidemia from a Brazilian National Surveillance Program. PLoS One 2016, 11, e0146909,  DOI: 10.1371/journal.pone.0146909
  12. 12
    Chowdhary, A.; Anil Kumar, V.; Sharma, C.; Prakash, A.; Agarwal, K.; Babu, R.; Dinesh, K. R.; Karim, S.; Singh, S. K.; Hagen, F.; Meis, J. F. Multidrug-resistant endemic clonal strain of Candida auris in India. Eur. J. Clin. Microbiol. Infect. Dis. 2014, 33, 919926,  DOI: 10.1007/s10096-013-2027-1
  13. 13
    Morales-López, S. E.; Parra-Giraldo, C. M.; Ceballos-Garzon, A.; Martinez, H. P.; Rodriguez, G. J.; Alvarez-Moreno, C. A.; Rodriguez, J. Y. Invasive Infections with Multidrug-Resistant Yeast Candida auris, Colombia. Emerg. Infect. Dis. 2017, 23, 162164,  DOI: 10.3201/eid2301.161497
  14. 14
    Sayeed, M. A.; Farooqi, J.; Jabeen, K.; Awan, S.; Mahmood, S. F. Clinical spectrum and factors impacting outcome of Candida auris: a single center study from Pakistan. BMC Infect. Dis. 2019, 19, 384,  DOI: 10.1186/s12879-019-3999-y
  15. 15
    Al-Siyabi, T.; Al Busaidi, I.; Balkhair, A.; Al-Muharrmi, Z.; Al-Salti, M.; Al’Adawi, B. First report of Candida auris in Oman: Clinical and microbiological description of five candidemia cases. J. Infect. 2017, 75, 373376,  DOI: 10.1016/j.jinf.2017.05.016
  16. 16
    CDC Tracking Candida auris. https://www.cdc.gov/fungal/candida-auris/tracking-c-auris.html (accessed August 12, 2021).
  17. 17
    Morton, J. F. Brazilian pepper─Its impact on people, animals and the environment. Econ Bot 1978, 32, 353359,  DOI: 10.1007/BF02907927
  18. 18
    Dettweiler, M.; Marquez, L.; Lin, M.; Sweeney-Jones, A. M.; Chhetri, B. K.; Zurawski, D. V.; Kubanek, J.; Quave, C. L. Pentagalloyl glucose from Schinus terebinthifolia inhibits growth of carbapenem-resistant Acinetobacter baumannii. Sci. Rep. 2020, 10, 15340  DOI: 10.1038/s41598-020-72331-w
  19. 19
    Cho, J.-Y.; Sohn, M.-J.; Lee, J.; Kim, W.-G. Isolation and identification of pentagalloylglucose with broad-spectrum antibacterial activity from Rhus trichocarpa Miquel. Food Chem. 2010, 123, 501506,  DOI: 10.1016/j.foodchem.2010.04.072
  20. 20
    Oh, G.-S.; Pae, H.-O.; Oh, H.; Hong, S.-G.; Kim, I.-K.; Chai, K.-Y.; Yun, Y.-G.; Kwon, T.-O.; Chung, H.-T. In vitro anti-proliferative effect of 1,2,3,4,6-penta-O-galloyl-beta-d-glucose on human hepatocellular carcinoma cell line, SK-HEP-1 cells. Cancer Lett. 2001, 174, 1724,  DOI: 10.1016/S0304-3835(01)00680-2
  21. 21
    Shaikh, Q.-u.-a.; Yang, M.; Memon, K. H.; Lateef, M.; Na, D.; Wan, S.; Eric, D.; Zhang, L.; Jiang, T. 1,2,3,4,6-Pentakis[-O-(3,4,5-trihydroxybenzoyl)]-α,β-D-glucopyranose (PGG) analogs: design, synthesis, anti-tumor and anti-oxidant activities. Carbohydr. Res. 2016, 430, 7281,  DOI: 10.1016/j.carres.2016.04.021
  22. 22
    Kawk, S. H.; Kang, Y. R.; Kim, Y. H. 1,2,3,4,6-Penta-O-galloyl-β-d-glucose suppresses colon cancer through induction of tumor suppressor. Bioorg. Med. Chem. Lett. 2018, 28, 21172123,  DOI: 10.1016/j.bmcl.2018.05.028
  23. 23
    Mendonca, P.; Alghamdi, S.; Messeha, S.; Soliman, K. F. A. Pentagalloyl glucose inhibits TNF-α-activated CXCL1/GRO-α expression and induces apoptosis-related genes in triple-negative breast cancer cells. Sci. Rep. 2021, 11, 5649  DOI: 10.1038/s41598-021-85090-z
  24. 24
    Joo, Y.-H.; Lee, Y.-G.; Lim, Y.; Jeon, H.; Kim, E. H.; Choi, J.; Hong, W.; Jeon, H.; Ahrweiler, M.; Kim, H.; Kang, S. C.; Seo, Y.-J. Potent antiviral activity of the extract of Elaeocarpus sylvestris against influenza A virus in vitro and in vivo. Phytomedicine 2022, 97, 153892  DOI: 10.1016/j.phymed.2021.153892
  25. 25
    CDC Candida Auris: Antifungal Susceptibility Testing and Interpretation. https://www.cdc.gov/fungal/candida-auris/c-auris-antifungal.html, (accessed November 20, 2022).
  26. 26
    Calabrese, D.; Bille, J.; Sanglard, D. A novel multidrug efflux transporter gene of the major facilitator superfamily from Candida albicans (FLU1) conferring resistance to fluconazole. Microbiology 2000, 146, 27432754,  DOI: 10.1099/00221287-146-11-2743
  27. 27
    Ryan, P.; Hynes, M. J. The kinetics and mechanisms of the complex formation and antioxidant behaviour of the polyphenols EGCg and ECG with iron(III). J. Inorg. Biochem. 2007, 101, 585593,  DOI: 10.1016/j.jinorgbio.2006.12.001
  28. 28
    Jones, M. M.; Innes, K. K. Restrictions on the Use of Job’s Method. J. Phys. Chem. A 1958, 62, 10051008,  DOI: 10.1021/j150566a031
  29. 29
    Vosburgh, W. C.; Cooper, G. R. Complex Ions. I. The Identification of Complex Ions in Solution by Spectrophotometric Measurements. J. Am. Chem. Soc. 1941, 63, 437442,  DOI: 10.1021/ja01847a025
  30. 30
    Baek, Y. U.; Li, M.; Davis, D. A. Candida albicans ferric reductases are differentially regulated in response to distinct forms of iron limitation by the Rim101 and CBF transcription factors. Eukaryot Cell 2008, 7, 11681179,  DOI: 10.1128/EC.00108-08
  31. 31
    Hsu, P. C.; Yang, C. Y.; Lan, C. Y. Candida albicans Hap43 is a repressor induced under low-iron conditions and is essential for iron-responsive transcriptional regulation and virulence. Eukaryot Cell 2011, 10, 207225,  DOI: 10.1128/EC.00158-10
  32. 32
    Ramírez-Zavala, B.; Krüger, I.; Dunker, C.; Jacobsen, I. D.; Morschhäuser, J. The protein kinase Ire1 has a Hac1-independent essential role in iron uptake and virulence of Candida albicans. PLoS Pathog. 2022, 18, e1010283  DOI: 10.1371/journal.ppat.1010283
  33. 33
    Cryan, L. M.; Bazinet, L.; Habeshian, K. A.; Cao, S.; Clardy, J.; Christensen, K. A.; Rogers, M. S. 1,2,3,4,6-Penta-O-galloyl-beta-D-glucopyranose inhibits angiogenesis via inhibition of capillary morphogenesis gene 2. J. Med. Chem. 2013, 56, 19401945,  DOI: 10.1021/jm301558t
  34. 34
    Feldman, K. S.; Sahasrabudhe, K.; Lawlor, M. D.; Wilson, S. L.; Lang, C. H.; Scheuchenzuber, W. J. In vitro and In vivo inhibition of LPS-stimulated tumor necrosis factor-alpha secretion by the gallotannin beta-D-pentagalloylglucose. Bioorg. Med. Chem. Lett. 2001, 11, 18131815,  DOI: 10.1016/S0960-894X(01)00332-8
  35. 35
    Cheng, S. W. K.; Eagleton, M.; Echeverri, S.; Munoz, J. G.; Holden, A. H.; Hill, A. A.; Krievins, D.; Ramaiah, V. A pilot study to evaluate a novel localized treatment to stabilize small- to medium-sized infrarenal abdominal aortic aneurysms. J. Vasc. Surg. 2023,  DOI: 10.1016/j.jvs.2023.05.056
  36. 36
    Jiamboonsri, P.; Pithayanukul, P.; Bavovada, R.; Leanpolchareanchai, J.; Yin, T.; Gao, S.; Hu, M. Factors Influencing Oral Bioavailability of Thai Mango Seed Kernel Extract and Its Key Phenolic Principles. Molecules 2015, 20, 2125421273,  DOI: 10.3390/molecules201219759
  37. 37
    Li, L.; Shaik, A. A.; Zhang, J.; Nhkata, K.; Wang, L.; Zhang, Y.; Xing, C.; Kim, S. H.; Lu, J. Preparation of penta-O-galloyl-beta-D-glucose from tannic acid and plasma pharmacokinetic analyses by liquid-liquid extraction and reverse-phase HPLC. J. Pharm. Biomed. Anal. 2011, 54, 545550,  DOI: 10.1016/j.jpba.2010.09.028
  38. 38
    Ma, C.-x.; Zhao, X.; Wang, P.; Jia, P.; Zhao, X.-f.; Xiao, C.-n.; Zheng, X.-h. Metabolite characterization of Penta-O-galloyl-β-D-glucose in rat biofluids by HPLC-QTOF-MS. Chin. Herb. Med. 2018, 10, 7379,  DOI: 10.1016/j.chmed.2018.01.002
  39. 39
    Hatcher, H. C.; Singh, R. N.; Torti, F. M.; Torti, S. V. Synthetic and natural iron chelators: therapeutic potential and clinical use. Future Med. Chem. 2009, 1, 16431670,  DOI: 10.4155/fmc.09.121
  40. 40
    Bairwa, G.; Hee Jung, W.; Kronstad, J. W. Iron acquisition in fungal pathogens of humans. Metallomics 2017, 9, 215227,  DOI: 10.1039/C6MT00301J
  41. 41
    Gerwien, F.; Skrahina, V.; Kasper, L.; Hube, B.; Brunke, S. Metals in fungal virulence. FEMS Microbiol. Rev. 2018, 42, fux050,  DOI: 10.1093/femsre/fux050
  42. 42
    Fourie, R.; Kuloyo, O. O.; Mochochoko, B. M.; Albertyn, J.; Pohl, C. H. Iron at the Centre of Candida albicans Interactions. Front. Cell. Infect. Microbiol. 2018, 8, 185,  DOI: 10.3389/fcimb.2018.00185
  43. 43
    Dittmar, W.; Lohaus, G. HOE 296, a new antimycotic compound with a broad antimicrobial spectrum. Laboratory results. Arzneimittelforschung 1973, 23, 670674
  44. 44
    Subissi, A.; Monti, D.; Togni, G.; Mailland, F. Ciclopirox. Drugs 2010, 70, 21332152,  DOI: 10.2165/11538110-000000000-00000
  45. 45
    Niewerth, M.; Kunze, D.; Seibold, M.; Schaller, M.; Korting, H. C.; Hube, B. Ciclopirox olamine treatment affects the expression pattern of Candida albicans genes encoding virulence factors, iron metabolism proteins, and drug resistance factors. Antimicrob. Agents Chemother. 2003, 47, 18051817,  DOI: 10.1128/AAC.47.6.1805-1817.2003
  46. 46
    Lee, R. E. B.; Liu, T. T.; Barker, K. S.; Lee, R. E.; Rogers, P. D. Genome-wide expression profiling of the response to ciclopirox olamine in Candida albicans. J. Antimicrob. Chemother 2005, 55, 655662,  DOI: 10.1093/jac/dki105
  47. 47
    Sigle, H.-C.; Thewes, S.; Niewerth, M.; Korting, H. C.; Schäfer-Korting, M.; Hube, B. Oxygen accessibility and iron levels are critical factors for the antifungal action of ciclopirox against Candida albicans. J. Antimicrob. Chemother 2005, 55, 663673,  DOI: 10.1093/jac/dki089
  48. 48
    Linden, T.; Katschinski, D. M.; Eckhardt, K.; Scheid, A.; Pagel, H.; Wenger, R. H. The antimycotic ciclopirox olamine induces HIF-1α stability, VEGF expression, and angiogenesis. FASEB J. 2003, 17, 761763,  DOI: 10.1096/fj.02-0586fje
  49. 49
    Skrzypek, M. S.; Binkley, J.; Binkley, G.; Miyasato, S. R.; Simison, M.; Sherlock, G. Candida Genome Database: C. albicans HAP43/C1_11210C.. http://www.candidagenome.org/cgi-bin/locus.pl?dbid=CAL0000188360, (accessed 06-27-2023).
  50. 50
    Zinatizadeh, M. R.; Schock, B.; Chalbatani, G. M.; Zarandi, P. K.; Jalali, S. A.; Miri, S. R. The Nuclear Factor Kappa B (NF-kB) signaling in cancer development and immune diseases. Genes Dis. 2021, 8, 287297,  DOI: 10.1016/j.gendis.2020.06.005
  51. 51
    Xiong, S.; She, H.; Takeuchi, H.; Han, B.; Engelhardt, J. F.; Barton, C. H.; Zandi, E.; Giulivi, C.; Tsukamoto, H. Signaling Role of Intracellular Iron in NF-κB Activation*. J. Biol. Chem. 2003, 278, 1764617654,  DOI: 10.1074/jbc.M210905200
  52. 52
    She, H.; Xiong, S.; Lin, M.; Zandi, E.; Giulivi, C.; Tsukamoto, H. Iron activates NF-κB in Kupffer cells. Am. J. Physiol.: Gastrointest. Liver Physiol. 2002, 283, G719G726,  DOI: 10.1152/ajpgi.00108.2002
  53. 53
    Kim, B. M.; Choi, J. Y.; Kim, Y. J.; Woo, H. D.; Chung, H. W. Desferrioxamine (DFX) has genotoxic effects on cultured human lymphocytes and induces the p53-mediated damage response. Toxicology 2007, 229, 226235,  DOI: 10.1016/j.tox.2006.10.022
  54. 54
    Liang, S. X.; Richardson, D. R. The effect of potent iron chelators on the regulation of p53: examination of the expression, localization and DNA-binding activity of p53 and the transactivation of WAF1. Carcinogenesis 2003, 24, 16011614,  DOI: 10.1093/carcin/bgg116
  55. 55
    Torres-León, C.; Ventura-Sobrevilla, J.; Serna-Cock, L.; Ascacio-Valdés, J. A.; Contreras-Esquivel, J.; Aguilar, C. N. Pentagalloylglucose (PGG): A valuable phenolic compound with functional properties. J. Funct. Foods 2017, 37, 176189,  DOI: 10.1016/j.jff.2017.07.045
  56. 56
    Lee, Y.; Hossain, S.; MacAlpine, J.; Robbins, N.; Cowen, L. E. Functional genomic analysis of Candida albicans protein kinases reveals modulators of morphogenesis in diverse environments. iScience 2023, 26, 106145  DOI: 10.1016/j.isci.2023.106145
  57. 57
    Homann, O. R.; Dea, J.; Noble, S. M.; Johnson, A. D. A Phenotypic Profile of the Candida albicans Regulatory Network. PLoS Genet. 2009, 5, e1000783  DOI: 10.1371/journal.pgen.1000783
  58. 58
    Clinical and Laboratory Standards Institute, Performance standards for antimicrobial susceptibility testing: twenty-third informational supplement ; M100 - S23. CLSI: 2013.
  59. 59
    Odds, F. C. Synergy, antagonism, and what the chequerboard puts between them. J. Antimicrob. Chemother. 2003, 52, 1,  DOI: 10.1093/jac/dkg301
  60. 60
    Leite, M. C. A.; Bezerra, A. P. d. B.; Sousa, J. P. d.; Guerra, F. Q. S.; Lima, E. d. O. Evaluation of antifungal activity and mechanism of action of citral against Candida albicans. J. Evidence-Based Complementary Altern. Med. 2014, 2014, 378280  DOI: 10.1155/2014/378280
  61. 61
    Singh, S. D.; Robbins, N.; Zaas, A. K.; Schell, W. A.; Perfect, J. R.; Cowen, L. E. Hsp90 governs echinocandin resistance in the pathogenic yeast Candida albicans via calcineurin. PLoS Pathog. 2009, 5, e1000532  DOI: 10.1371/journal.ppat.1000532

Cited By

Click to copy section linkSection link copied!

This article has not yet been cited by other publications.

Open PDF

ACS Infectious Diseases

Cite this: ACS Infect. Dis. 2023, 9, 9, 1685–1694
Click to copy citationCitation copied!
https://doi.org/10.1021/acsinfecdis.3c00113
Published August 22, 2023

Copyright © 2023 The Authors. Published by American Chemical Society. This publication is licensed under

CC-BY 4.0 .

Article Views

3486

Altmetric

-

Citations

-
Learn about these metrics

Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.

Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.

The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.

  • Abstract

    Figure 1

    Figure 1. Chemical structure of penta-O-galloyl-β-d-glucose, PGG.

    Figure 2

    Figure 2. Penta-O-galloyl-β-d-glucose displays broad spectrum anti-Candida activity. Growth inhibition of PGG against (A) C. albicans, (B) C. glabrata, (C) C. parapsilosis, and (D) C. auris. Solid lines represent PGG. Dashed lines represent the positive control fluconazole. The data are plotted as the mean % growth inhibition ± SD as compared to the DMSO vehicle from two independent experiments. The horizontal dotted line represents 90% growth inhibition. See Supplemental Figures S8–S11 for expanded graphs including additional positive controls. See Table 2 for strain details.

    Figure 3

    Figure 3. PGG sensitivity in C. albicans is not modulated by efflux pumps. The cdr1Δ/Δ, cdr2Δ/Δ, mdr1Δ/Δ, flu1Δ/Δ, and wild-type strains were grown overnight in YPD and subject to dose–response assays in RPMI-1640 medium with twofold dilution gradients of PGG. Fluconazole was included as a positive control. Growth (OD600nm) was measured after 72 h of incubation at 30 °C, averaged between technical duplicates, and normalized to the drug-free wild-type control. Results are presented in a heat-map format with a scale bar provided at the bottom of the figure. All assays were performed in biological duplicates.

    Figure 4

    Figure 4. Growth inhibition of PGG against multiple Candida species is abrogated by iron supplementation, and PGG binds multiple iron ions. (A) C. albicans 90028, (B) C. glabrata CDC325, and (C) C. auris CDC381 were treated with PGG or Amphotericin B at varying concentrations (4, 8, and 16 μg/mL) and media was supplemented with a 10 mM solution of ferrous sulfate (Fe2+) or ferric sulfate (Fe3+) and % growth inhibition measured after 48 h. The data are plotted as the mean % growth inhibition ± SD as compared to the DMSO vehicle from two independent experiments. The horizontal dotted line represents 90% growth inhibition compared to the vehicle. Statistical analysis was done using two-way ANOVA with multiple comparisons. (D) PGG or EGCG was allowed to react with varying concentrations of iron (III) in solution at pH 2.0. The bipy solution was added to the mixtures and the formation of the [Fe(bipy)3]2+ complex was measured at 520 nm. [PGG]total, [EGCG]total = 10 μM. The data are plotted as the mean absorbance ± SD from two independent experiments.

    Figure 5

    Figure 5. Iron homeostasis genes are required for PGG tolerance in C. albicans. The ire1Δ/ire1Δ, hap43Δ/hap43Δ, and rim101Δ/rim101Δ strains and their respective wild-type controls were grown overnight in YPD and subject to dose–response assays in RPMI-1640 medium with twofold dilution gradients of PGG. The metal chelator BPS was included as a positive control. After 48 h incubation at 30 °C, the relative viable cell number was assessed. Data were averaged between technical duplicates and normalized to wild-type drug-free control wells. Results are presented in a heat-map format with a color scale bar presented at the bottom of the figure. All assays were performed in biological duplicates.

  • References


    This article references 61 other publications.

    1. 1
      Limon, J. J.; Skalski, J. H.; Underhill, D. M. Commensal Fungi in Health and Disease. Cell Host Microbe 2017, 22, 156165,  DOI: 10.1016/j.chom.2017.07.002
    2. 2
      Neville, B. A.; d’Enfert, C.; Bougnoux, M.-E. Candida albicans commensalism in the gastrointestinal tract. FEMS Yeast Res 2015, 15, fov081,  DOI: 10.1093/femsyr/fov081
    3. 3
      Bilal, H.; Hou, B.; Shafiq, M.; Chen, X.; Shahid, M. A.; Zeng, Y. Antifungal susceptibility pattern of Candida isolated from cutaneous candidiasis patients in eastern Guangdong region: A retrospective study of the past 10 years. Front. Microbiol. 2022, 13, 981181  DOI: 10.3389/fmicb.2022.981181
    4. 4
      Khodadadi, H.; Zomorodian, K.; Nouraei, H.; Zareshahrabadi, Z.; Barzegar, S.; Zare, M. R.; Pakshir, K. Prevalence of superficial-cutaneous fungal infections in Shiraz, Iran: A five-year retrospective study (2015–2019). J. Clin. Lab. Anal. 2021, 35, e23850  DOI: 10.1002/jcla.23850
    5. 5
      Otašević, S.; Momčilović, S.; Golubović, M.; Ignjatović, A.; Rančić, N.; Đorđević, M.; Ranđelović, M.; Hay, R.; Arsić-Arsenijević, V. Species distribution and epidemiological characteristics of superficial fungal infections in Southeastern Serbia. Mycoses 2019, 62, 458465,  DOI: 10.1111/myc.12900
    6. 6
      Pappas, P. G.; Lionakis, M. S.; Arendrup, M. C.; Ostrosky-Zeichner, L.; Kullberg, B. J. Invasive candidiasis. Nat. Rev. Dis. Primers 2018, 4, 18026,  DOI: 10.1038/nrdp.2018.26
    7. 7
      Tsay, S.; Williams, S.; Mu, Y.; Epson, E.; Johnston, H.; Farley, M. M.; Harrison, L. H.; Vonbank, B.; Shrum, S.; Dumyati, G.; Zhang, A.; Schaffner, W.; Magill, S.; Vallabhaneni, S. 363. National Burden of Candidemia, United States, 2017. Open Forum Infect. Dis. 2018, 5, S142S143,  DOI: 10.1093/ofid/ofy210.374
    8. 8
      Puig-Asensio, M.; Padilla, B.; Garnacho-Montero, J.; Zaragoza, O.; Aguado, J. M.; Zaragoza, R.; Montejo, M.; Muñoz, P.; Ruiz-Camps, I.; Cuenca-Estrella, M.; Almirante, B. Epidemiology and predictive factors for early and late mortality in Candida bloodstream infections: a population-based surveillance in Spain. Clin. Microbiol. Infect. 2014, 20, O24554,  DOI: 10.1111/1469-0691.12380
    9. 9
      Cleveland, A. A.; Farley, M. M.; Harrison, L. H.; Stein, B.; Hollick, R.; Lockhart, S. R.; Magill, S. S.; Derado, G.; Park, B. J.; Chiller, T. M. Changes in incidence and antifungal drug resistance in candidemia: results from population-based laboratory surveillance in Atlanta and Baltimore, 2008-2011. Clin. Infect. Dis. 2012, 55, 13521361,  DOI: 10.1093/cid/cis697
    10. 10
      Kreusch, A.; Karstaedt, A. S. Candidemia among adults in Soweto, South Africa, 1990-2007. Int. J. Infect. Dis. 2013, 17, e6213,  DOI: 10.1016/j.ijid.2013.02.010
    11. 11
      Doi, A. M.; Pignatari, A. C. C.; Edmond, M. B.; Marra, A. R.; Camargo, L. F. A.; Siqueira, R. A.; da Mota, V. P.; Colombo, A. L. Epidemiology and Microbiologic Characterization of Nosocomial Candidemia from a Brazilian National Surveillance Program. PLoS One 2016, 11, e0146909,  DOI: 10.1371/journal.pone.0146909
    12. 12
      Chowdhary, A.; Anil Kumar, V.; Sharma, C.; Prakash, A.; Agarwal, K.; Babu, R.; Dinesh, K. R.; Karim, S.; Singh, S. K.; Hagen, F.; Meis, J. F. Multidrug-resistant endemic clonal strain of Candida auris in India. Eur. J. Clin. Microbiol. Infect. Dis. 2014, 33, 919926,  DOI: 10.1007/s10096-013-2027-1
    13. 13
      Morales-López, S. E.; Parra-Giraldo, C. M.; Ceballos-Garzon, A.; Martinez, H. P.; Rodriguez, G. J.; Alvarez-Moreno, C. A.; Rodriguez, J. Y. Invasive Infections with Multidrug-Resistant Yeast Candida auris, Colombia. Emerg. Infect. Dis. 2017, 23, 162164,  DOI: 10.3201/eid2301.161497
    14. 14
      Sayeed, M. A.; Farooqi, J.; Jabeen, K.; Awan, S.; Mahmood, S. F. Clinical spectrum and factors impacting outcome of Candida auris: a single center study from Pakistan. BMC Infect. Dis. 2019, 19, 384,  DOI: 10.1186/s12879-019-3999-y
    15. 15
      Al-Siyabi, T.; Al Busaidi, I.; Balkhair, A.; Al-Muharrmi, Z.; Al-Salti, M.; Al’Adawi, B. First report of Candida auris in Oman: Clinical and microbiological description of five candidemia cases. J. Infect. 2017, 75, 373376,  DOI: 10.1016/j.jinf.2017.05.016
    16. 16
      CDC Tracking Candida auris. https://www.cdc.gov/fungal/candida-auris/tracking-c-auris.html (accessed August 12, 2021).
    17. 17
      Morton, J. F. Brazilian pepper─Its impact on people, animals and the environment. Econ Bot 1978, 32, 353359,  DOI: 10.1007/BF02907927
    18. 18
      Dettweiler, M.; Marquez, L.; Lin, M.; Sweeney-Jones, A. M.; Chhetri, B. K.; Zurawski, D. V.; Kubanek, J.; Quave, C. L. Pentagalloyl glucose from Schinus terebinthifolia inhibits growth of carbapenem-resistant Acinetobacter baumannii. Sci. Rep. 2020, 10, 15340  DOI: 10.1038/s41598-020-72331-w
    19. 19
      Cho, J.-Y.; Sohn, M.-J.; Lee, J.; Kim, W.-G. Isolation and identification of pentagalloylglucose with broad-spectrum antibacterial activity from Rhus trichocarpa Miquel. Food Chem. 2010, 123, 501506,  DOI: 10.1016/j.foodchem.2010.04.072
    20. 20
      Oh, G.-S.; Pae, H.-O.; Oh, H.; Hong, S.-G.; Kim, I.-K.; Chai, K.-Y.; Yun, Y.-G.; Kwon, T.-O.; Chung, H.-T. In vitro anti-proliferative effect of 1,2,3,4,6-penta-O-galloyl-beta-d-glucose on human hepatocellular carcinoma cell line, SK-HEP-1 cells. Cancer Lett. 2001, 174, 1724,  DOI: 10.1016/S0304-3835(01)00680-2
    21. 21
      Shaikh, Q.-u.-a.; Yang, M.; Memon, K. H.; Lateef, M.; Na, D.; Wan, S.; Eric, D.; Zhang, L.; Jiang, T. 1,2,3,4,6-Pentakis[-O-(3,4,5-trihydroxybenzoyl)]-α,β-D-glucopyranose (PGG) analogs: design, synthesis, anti-tumor and anti-oxidant activities. Carbohydr. Res. 2016, 430, 7281,  DOI: 10.1016/j.carres.2016.04.021
    22. 22
      Kawk, S. H.; Kang, Y. R.; Kim, Y. H. 1,2,3,4,6-Penta-O-galloyl-β-d-glucose suppresses colon cancer through induction of tumor suppressor. Bioorg. Med. Chem. Lett. 2018, 28, 21172123,  DOI: 10.1016/j.bmcl.2018.05.028
    23. 23
      Mendonca, P.; Alghamdi, S.; Messeha, S.; Soliman, K. F. A. Pentagalloyl glucose inhibits TNF-α-activated CXCL1/GRO-α expression and induces apoptosis-related genes in triple-negative breast cancer cells. Sci. Rep. 2021, 11, 5649  DOI: 10.1038/s41598-021-85090-z
    24. 24
      Joo, Y.-H.; Lee, Y.-G.; Lim, Y.; Jeon, H.; Kim, E. H.; Choi, J.; Hong, W.; Jeon, H.; Ahrweiler, M.; Kim, H.; Kang, S. C.; Seo, Y.-J. Potent antiviral activity of the extract of Elaeocarpus sylvestris against influenza A virus in vitro and in vivo. Phytomedicine 2022, 97, 153892  DOI: 10.1016/j.phymed.2021.153892
    25. 25
      CDC Candida Auris: Antifungal Susceptibility Testing and Interpretation. https://www.cdc.gov/fungal/candida-auris/c-auris-antifungal.html, (accessed November 20, 2022).
    26. 26
      Calabrese, D.; Bille, J.; Sanglard, D. A novel multidrug efflux transporter gene of the major facilitator superfamily from Candida albicans (FLU1) conferring resistance to fluconazole. Microbiology 2000, 146, 27432754,  DOI: 10.1099/00221287-146-11-2743
    27. 27
      Ryan, P.; Hynes, M. J. The kinetics and mechanisms of the complex formation and antioxidant behaviour of the polyphenols EGCg and ECG with iron(III). J. Inorg. Biochem. 2007, 101, 585593,  DOI: 10.1016/j.jinorgbio.2006.12.001
    28. 28
      Jones, M. M.; Innes, K. K. Restrictions on the Use of Job’s Method. J. Phys. Chem. A 1958, 62, 10051008,  DOI: 10.1021/j150566a031
    29. 29
      Vosburgh, W. C.; Cooper, G. R. Complex Ions. I. The Identification of Complex Ions in Solution by Spectrophotometric Measurements. J. Am. Chem. Soc. 1941, 63, 437442,  DOI: 10.1021/ja01847a025
    30. 30
      Baek, Y. U.; Li, M.; Davis, D. A. Candida albicans ferric reductases are differentially regulated in response to distinct forms of iron limitation by the Rim101 and CBF transcription factors. Eukaryot Cell 2008, 7, 11681179,  DOI: 10.1128/EC.00108-08
    31. 31
      Hsu, P. C.; Yang, C. Y.; Lan, C. Y. Candida albicans Hap43 is a repressor induced under low-iron conditions and is essential for iron-responsive transcriptional regulation and virulence. Eukaryot Cell 2011, 10, 207225,  DOI: 10.1128/EC.00158-10
    32. 32
      Ramírez-Zavala, B.; Krüger, I.; Dunker, C.; Jacobsen, I. D.; Morschhäuser, J. The protein kinase Ire1 has a Hac1-independent essential role in iron uptake and virulence of Candida albicans. PLoS Pathog. 2022, 18, e1010283  DOI: 10.1371/journal.ppat.1010283
    33. 33
      Cryan, L. M.; Bazinet, L.; Habeshian, K. A.; Cao, S.; Clardy, J.; Christensen, K. A.; Rogers, M. S. 1,2,3,4,6-Penta-O-galloyl-beta-D-glucopyranose inhibits angiogenesis via inhibition of capillary morphogenesis gene 2. J. Med. Chem. 2013, 56, 19401945,  DOI: 10.1021/jm301558t
    34. 34
      Feldman, K. S.; Sahasrabudhe, K.; Lawlor, M. D.; Wilson, S. L.; Lang, C. H.; Scheuchenzuber, W. J. In vitro and In vivo inhibition of LPS-stimulated tumor necrosis factor-alpha secretion by the gallotannin beta-D-pentagalloylglucose. Bioorg. Med. Chem. Lett. 2001, 11, 18131815,  DOI: 10.1016/S0960-894X(01)00332-8
    35. 35
      Cheng, S. W. K.; Eagleton, M.; Echeverri, S.; Munoz, J. G.; Holden, A. H.; Hill, A. A.; Krievins, D.; Ramaiah, V. A pilot study to evaluate a novel localized treatment to stabilize small- to medium-sized infrarenal abdominal aortic aneurysms. J. Vasc. Surg. 2023,  DOI: 10.1016/j.jvs.2023.05.056
    36. 36
      Jiamboonsri, P.; Pithayanukul, P.; Bavovada, R.; Leanpolchareanchai, J.; Yin, T.; Gao, S.; Hu, M. Factors Influencing Oral Bioavailability of Thai Mango Seed Kernel Extract and Its Key Phenolic Principles. Molecules 2015, 20, 2125421273,  DOI: 10.3390/molecules201219759
    37. 37
      Li, L.; Shaik, A. A.; Zhang, J.; Nhkata, K.; Wang, L.; Zhang, Y.; Xing, C.; Kim, S. H.; Lu, J. Preparation of penta-O-galloyl-beta-D-glucose from tannic acid and plasma pharmacokinetic analyses by liquid-liquid extraction and reverse-phase HPLC. J. Pharm. Biomed. Anal. 2011, 54, 545550,  DOI: 10.1016/j.jpba.2010.09.028
    38. 38
      Ma, C.-x.; Zhao, X.; Wang, P.; Jia, P.; Zhao, X.-f.; Xiao, C.-n.; Zheng, X.-h. Metabolite characterization of Penta-O-galloyl-β-D-glucose in rat biofluids by HPLC-QTOF-MS. Chin. Herb. Med. 2018, 10, 7379,  DOI: 10.1016/j.chmed.2018.01.002
    39. 39
      Hatcher, H. C.; Singh, R. N.; Torti, F. M.; Torti, S. V. Synthetic and natural iron chelators: therapeutic potential and clinical use. Future Med. Chem. 2009, 1, 16431670,  DOI: 10.4155/fmc.09.121
    40. 40
      Bairwa, G.; Hee Jung, W.; Kronstad, J. W. Iron acquisition in fungal pathogens of humans. Metallomics 2017, 9, 215227,  DOI: 10.1039/C6MT00301J
    41. 41
      Gerwien, F.; Skrahina, V.; Kasper, L.; Hube, B.; Brunke, S. Metals in fungal virulence. FEMS Microbiol. Rev. 2018, 42, fux050,  DOI: 10.1093/femsre/fux050
    42. 42
      Fourie, R.; Kuloyo, O. O.; Mochochoko, B. M.; Albertyn, J.; Pohl, C. H. Iron at the Centre of Candida albicans Interactions. Front. Cell. Infect. Microbiol. 2018, 8, 185,  DOI: 10.3389/fcimb.2018.00185
    43. 43
      Dittmar, W.; Lohaus, G. HOE 296, a new antimycotic compound with a broad antimicrobial spectrum. Laboratory results. Arzneimittelforschung 1973, 23, 670674
    44. 44
      Subissi, A.; Monti, D.; Togni, G.; Mailland, F. Ciclopirox. Drugs 2010, 70, 21332152,  DOI: 10.2165/11538110-000000000-00000
    45. 45
      Niewerth, M.; Kunze, D.; Seibold, M.; Schaller, M.; Korting, H. C.; Hube, B. Ciclopirox olamine treatment affects the expression pattern of Candida albicans genes encoding virulence factors, iron metabolism proteins, and drug resistance factors. Antimicrob. Agents Chemother. 2003, 47, 18051817,  DOI: 10.1128/AAC.47.6.1805-1817.2003
    46. 46
      Lee, R. E. B.; Liu, T. T.; Barker, K. S.; Lee, R. E.; Rogers, P. D. Genome-wide expression profiling of the response to ciclopirox olamine in Candida albicans. J. Antimicrob. Chemother 2005, 55, 655662,  DOI: 10.1093/jac/dki105
    47. 47
      Sigle, H.-C.; Thewes, S.; Niewerth, M.; Korting, H. C.; Schäfer-Korting, M.; Hube, B. Oxygen accessibility and iron levels are critical factors for the antifungal action of ciclopirox against Candida albicans. J. Antimicrob. Chemother 2005, 55, 663673,  DOI: 10.1093/jac/dki089
    48. 48
      Linden, T.; Katschinski, D. M.; Eckhardt, K.; Scheid, A.; Pagel, H.; Wenger, R. H. The antimycotic ciclopirox olamine induces HIF-1α stability, VEGF expression, and angiogenesis. FASEB J. 2003, 17, 761763,  DOI: 10.1096/fj.02-0586fje
    49. 49
      Skrzypek, M. S.; Binkley, J.; Binkley, G.; Miyasato, S. R.; Simison, M.; Sherlock, G. Candida Genome Database: C. albicans HAP43/C1_11210C.. http://www.candidagenome.org/cgi-bin/locus.pl?dbid=CAL0000188360, (accessed 06-27-2023).
    50. 50
      Zinatizadeh, M. R.; Schock, B.; Chalbatani, G. M.; Zarandi, P. K.; Jalali, S. A.; Miri, S. R. The Nuclear Factor Kappa B (NF-kB) signaling in cancer development and immune diseases. Genes Dis. 2021, 8, 287297,  DOI: 10.1016/j.gendis.2020.06.005
    51. 51
      Xiong, S.; She, H.; Takeuchi, H.; Han, B.; Engelhardt, J. F.; Barton, C. H.; Zandi, E.; Giulivi, C.; Tsukamoto, H. Signaling Role of Intracellular Iron in NF-κB Activation*. J. Biol. Chem. 2003, 278, 1764617654,  DOI: 10.1074/jbc.M210905200
    52. 52
      She, H.; Xiong, S.; Lin, M.; Zandi, E.; Giulivi, C.; Tsukamoto, H. Iron activates NF-κB in Kupffer cells. Am. J. Physiol.: Gastrointest. Liver Physiol. 2002, 283, G719G726,  DOI: 10.1152/ajpgi.00108.2002
    53. 53
      Kim, B. M.; Choi, J. Y.; Kim, Y. J.; Woo, H. D.; Chung, H. W. Desferrioxamine (DFX) has genotoxic effects on cultured human lymphocytes and induces the p53-mediated damage response. Toxicology 2007, 229, 226235,  DOI: 10.1016/j.tox.2006.10.022
    54. 54
      Liang, S. X.; Richardson, D. R. The effect of potent iron chelators on the regulation of p53: examination of the expression, localization and DNA-binding activity of p53 and the transactivation of WAF1. Carcinogenesis 2003, 24, 16011614,  DOI: 10.1093/carcin/bgg116
    55. 55
      Torres-León, C.; Ventura-Sobrevilla, J.; Serna-Cock, L.; Ascacio-Valdés, J. A.; Contreras-Esquivel, J.; Aguilar, C. N. Pentagalloylglucose (PGG): A valuable phenolic compound with functional properties. J. Funct. Foods 2017, 37, 176189,  DOI: 10.1016/j.jff.2017.07.045
    56. 56
      Lee, Y.; Hossain, S.; MacAlpine, J.; Robbins, N.; Cowen, L. E. Functional genomic analysis of Candida albicans protein kinases reveals modulators of morphogenesis in diverse environments. iScience 2023, 26, 106145  DOI: 10.1016/j.isci.2023.106145
    57. 57
      Homann, O. R.; Dea, J.; Noble, S. M.; Johnson, A. D. A Phenotypic Profile of the Candida albicans Regulatory Network. PLoS Genet. 2009, 5, e1000783  DOI: 10.1371/journal.pgen.1000783
    58. 58
      Clinical and Laboratory Standards Institute, Performance standards for antimicrobial susceptibility testing: twenty-third informational supplement ; M100 - S23. CLSI: 2013.
    59. 59
      Odds, F. C. Synergy, antagonism, and what the chequerboard puts between them. J. Antimicrob. Chemother. 2003, 52, 1,  DOI: 10.1093/jac/dkg301
    60. 60
      Leite, M. C. A.; Bezerra, A. P. d. B.; Sousa, J. P. d.; Guerra, F. Q. S.; Lima, E. d. O. Evaluation of antifungal activity and mechanism of action of citral against Candida albicans. J. Evidence-Based Complementary Altern. Med. 2014, 2014, 378280  DOI: 10.1155/2014/378280
    61. 61
      Singh, S. D.; Robbins, N.; Zaas, A. K.; Schell, W. A.; Perfect, J. R.; Cowen, L. E. Hsp90 governs echinocandin resistance in the pathogenic yeast Candida albicans via calcineurin. PLoS Pathog. 2009, 5, e1000532  DOI: 10.1371/journal.ppat.1000532
  • Supporting Information

    Supporting Information


    The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsinfecdis.3c00113.

    • Experimental methods for concentration-response matrixes; effects of penta-O-galloyl-β-d-glucose upon human cytotoxicity and cell viability; media supplementation assays; Job’s method spectra; expanded MIC graphs with additional positive controls; and table with the additional Candida albicans strains and genotype (PDF)


    Terms & Conditions

    Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.