Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Int J Antimicrob Agents ; 62(3): 106906, 2023 Sep.
Article En | MEDLINE | ID: mdl-37392947

INTRODUCTION AND OBJECTIVES: The emergence of Candida auris has created a global health challenge. Azole antifungals are the most affected antifungal class because of the extraordinary capability of C. auris to develop resistance against these drugs. Here, we used a combinatorial therapeutic approach to sensitize C. auris to azole antifungals. METHODS AND RESULTS: We have demonstrated the capability of the HIV protease inhibitors lopinavir and ritonavir, at clinically relevant concentrations, to be used with azole antifungals to treat C. auris infections both in vitro and in vivo. Both lopinavir and ritonavir exhibited potent synergistic interactions with the azole antifungals, particularly with itraconazole against 24/24 (100%) and 31/34 (91%) of tested C. auris isolates, respectively. Furthermore, ritonavir significantly interfered with the fungal efflux pump, resulting in a significant increase in Nile red fluorescence by 44%. In a mouse model of C. auris systemic infection, ritonavir boosted the activity of lopinavir to work synergistically with fluconazole and itraconazole and significantly reduced the kidney fungal burden by a 1.2 log (∼94%) and 1.6 log (∼97%) CFU, respectively. CONCLUSION: Our results urge further comprehensive assessment of azoles and HIV protease inhibitors as a novel drug regimen for the treatment of serious invasive C. auris infections.


Candidiasis , HIV Protease Inhibitors , Animals , Mice , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Ritonavir/therapeutic use , Azoles/pharmacology , Azoles/therapeutic use , Itraconazole/pharmacology , Itraconazole/therapeutic use , Lopinavir/pharmacology , Lopinavir/therapeutic use , Candida auris , HIV Protease Inhibitors/pharmacology , HIV Protease Inhibitors/therapeutic use , Candidiasis/drug therapy , Candidiasis/microbiology , Microbial Sensitivity Tests , Drug Resistance, Fungal
2.
Anal Chem ; 95(26): 9901-9913, 2023 07 04.
Article En | MEDLINE | ID: mdl-37310727

Candida albicans (C. albicans), a major fungal pathogen, causes life-threatening infections in immunocompromised individuals. Fluconazole (FLC) is recommended as first-line therapy for treatment of invasive fungal infections. However, the widespread use of FLC has resulted in increased antifungal resistance among different strains of Candida, especially C. albicans, which is a leading source of hospital-acquired infections. Here, by hyperspectral stimulated Raman scattering imaging of single fungal cells in the fingerprint window and pixel-wise spectral unmixing, we report aberrant ergosteryl ester accumulation in azole-resistant C. albicans compared to azole-susceptible species. This accumulation was a consequence of de novo lipogenesis. Lipid profiling by mass spectroscopy identified ergosterol oleate to be the major species stored in azole-resistant C. albicans. Blocking ergosterol esterification by oleate and suppressing sterol synthesis by FLC synergistically suppressed the viability of C. albicans in vitro and limited the growth of biofilm on mouse skin in vivo. Our findings highlight a metabolic marker and a new therapeutic strategy for targeting azole-resistant C. albicans by interrupting the esterified ergosterol biosynthetic pathway.


Antifungal Agents , Candida albicans , Animals , Mice , Antifungal Agents/chemistry , Azoles/pharmacology , Azoles/metabolism , Spectrum Analysis, Raman , Esters/metabolism , Oleic Acid/metabolism , Microbial Sensitivity Tests , Fluconazole/metabolism , Ergosterol/pharmacology , Ergosterol/metabolism
3.
Front Cell Infect Microbiol ; 12: 1027394, 2022.
Article En | MEDLINE | ID: mdl-36275024

Tuberculosis, caused by Mycobacterium tuberculosis (Mtb) is an ancient disease that has remained a leading cause of infectious death. Mtb has evolved drug resistance to every antibiotic regimen ever introduced, greatly complicating treatment, lowering rates of cure and menacing TB control in parts of the world. As technology has advanced, our understanding of antimicrobial resistance has improved, and our models of the phenomenon have evolved. In this review, we focus on recent research progress that supports an updated model for the evolution of drug resistance in Mtb. We highlight the contribution of drug tolerance on the path to resistance, and the influence of heterogeneity on tolerance. Resistance is likely to remain an issue for as long as drugs are needed to treat TB. However, with technology driving new insights and careful management of newly developed resources, antimicrobial resistance need not continue to threaten global progress against TB, as it has done for decades.


Mycobacterium tuberculosis , Tuberculosis, Lymph Node , Humans , Mycobacterium tuberculosis/genetics , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Drug Resistance , Biology
4.
J Med Chem ; 65(9): 6612-6630, 2022 05 12.
Article En | MEDLINE | ID: mdl-35482444

Methicillin-resistant Staphylococcus aureus (MRSA) infections are still difficult to treat, despite the availability of many FDA-approved antibiotics. Thus, new compound scaffolds are still needed to treat MRSA. The oxadiazole-containing compound, HSGN-94, has been shown to reduce lipoteichoic acid (LTA) in S. aureus, but the mechanism that accounts for LTA biosynthesis inhibition remains uncharacterized. Herein, we report the elucidation of the mechanism by which HSGN-94 inhibits LTA biosynthesis via utilization of global proteomics, activity-based protein profiling, and lipid analysis via multiple reaction monitoring (MRM). Our data suggest that HSGN-94 inhibits LTA biosynthesis via direct binding to PgcA and downregulation of PgsA. We further show that HSGN-94 reduces the MRSA load in skin infection (mouse) and decreases pro-inflammatory cytokines in MRSA-infected wounds. Collectively, HSGN-94 merits further consideration as a potential drug for staphylococcal infections.


Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Animals , Anti-Bacterial Agents/chemistry , Mice , Microbial Sensitivity Tests , Oxadiazoles/metabolism , Oxadiazoles/pharmacology , Oxadiazoles/therapeutic use , Staphylococcal Infections/drug therapy , Staphylococcus aureus
6.
Virulence ; 11(1): 1466-1481, 2020 12.
Article En | MEDLINE | ID: mdl-33100149

With the rapid increase in the frequency of azole-resistant species, combination therapy appears to be a promising tool to augment the antifungal activity of azole drugs against resistant Candida species. Here, we report the effect of aprepitant, an antiemetic agent, on the antifungal activities of azole drugs against the multidrug-resistant Candida auris. Aprepitant reduced the minimum inhibitory concentration (MIC) of itraconazole in vitro, by up to eight-folds. Additionally, the aprepitant/itraconazole combination interfered significantly with the biofilm-forming ability of C. auris by 95 ± 0.13%, and significantly disrupted mature biofilms by 52 ± 0.83%, relative to the untreated control. In a Caenorhabditis elegans infection model, the aprepitant/itraconazole combination significantly prolonged the survival of infected nematodes by ~90% (five days post-infection) and reduced the fungal burden by ~92% relative to the untreated control. Further, this novel drug combination displayed broad-spectrum synergistic interactions against other medically important Candida species such as C. albicans, C. krusei, C. tropicalis, and C. parapsilosis (Æ©FICI ranged from 0.08 to 0.31). Comparative transcriptomic profiling and mechanistic studies indicated aprepitant/itraconazole interferes significantly with metal ion homeostasis and compromises the ROS detoxification ability of C. auris. This study presents aprepitant as a novel, potent, and broad-spectrum azole chemosensitizing agent that warrants further investigation.


Antiemetics/pharmacology , Antifungal Agents/pharmacology , Aprepitant/pharmacology , Azoles/pharmacology , Candida/drug effects , Animals , Biofilms/drug effects , Caenorhabditis elegans , Candida/physiology , Candidiasis/microbiology , Drug Resistance, Multiple, Fungal , Drug Synergism , Gene Expression Profiling , Homeostasis/drug effects , Ions , Metals , Microbial Sensitivity Tests
7.
Article En | MEDLINE | ID: mdl-33046487

The limited therapeutic options and the recent emergence of multidrug-resistant Candida species present a significant challenge to human medicine and underscore the need for novel therapeutic approaches. Drug repurposing appears as a promising tool to augment the activity of current azole antifungals, especially against multidrug-resistant Candida auris In this study, we evaluated the fluconazole chemosensitization activities of 1,547 FDA-approved drugs and clinical molecules against azole-resistant C. auris This led to the discovery that lopinavir, an HIV protease inhibitor, is a potent agent capable of sensitizing C. auris to the effect of azole antifungals. At a therapeutically achievable concentration, lopinavir exhibited potent synergistic interactions with azole drugs, particularly with itraconazole against C. auris (fractional inhibitory concentration index [ΣFICI] ranged from 0.04 to 0.09). Additionally, the lopinavir/itraconazole combination enhanced the survival rate of C. auris-infected Caenorhabditis elegans by 90% and reduced the fungal burden in infected nematodes by 88.5% (P < 0.05) relative to that of the untreated control. Furthermore, lopinavir enhanced the antifungal activity of itraconazole against other medically important Candida species, including C. albicans, C. tropicalis, C. krusei, and C. parapsilosis Comparative transcriptomic profiling and mechanistic studies revealed that lopinavir was able to significantly interfere with the glucose permeation and ATP synthesis. This compromised the efflux ability of C. auris and consequently enhanced the susceptibility to azole drugs, as demonstrated by Nile red efflux assays. Altogether, these findings present lopinavir as a novel, potent, and broad-spectrum azole-chemosensitizing agent that warrants further investigation against recalcitrant Candida infections.


Antifungal Agents , Pharmaceutical Preparations , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Azoles/pharmacology , Candida , Fluconazole , Humans , Lopinavir/pharmacology , Microbial Sensitivity Tests
8.
Biomaterials ; 262: 120344, 2020 12.
Article En | MEDLINE | ID: mdl-32905902

Bacterial pathogens residing in host macrophages in intracellular infections are hard to eradicate because traditional antibiotics do not readily enter the cells or get eliminated via efflux pumps. To overcome this challenge, we developed a new particle formulation with a size amenable to selective macrophage uptake, loaded with two antibacterial agents - pexiganan and silver (Ag) nanoparticles. Here, pexiganan was loaded in 600 nm poly(lactic-co-glycolic acid) (PLGA) particles (NP), and the particle surface was modified with an iron-tannic acid supramolecular complex (pTA) that help attach Ag nanoparticles. PLGA particles coated with Ag (NP-pTA-Ag) were taken up by macrophages, but not by non-phagocytic cells, such as fibroblasts, reducing non-specific toxicity associated with Ag nanoparticles. NP-pTA-Ag loaded with pexiganan (Pex@NP-pTA-Ag) showed more potent antibacterial activity against various intracellular pathogens than NP-pTA-Ag or Pex@NP (pexiganan-loaded NP with no Ag), suggesting a collaborative function between pexiganan and Ag nanoparticles. Mouse whole-body imaging demonstrated that, upon intravenous injection, NP-pTA-Ag quickly accumulated in the liver and spleen, where intracellular bacteria tend to reside. These results support that Pex@NP-pTA-Ag is a promising strategy for the treatment of intracellular bacterial infection.


Bacterial Infections , Metal Nanoparticles , Nanoparticles , Animals , Anti-Bacterial Agents/pharmacology , Macrophages , Mice , Silver
9.
J Org Chem ; 85(11): 7468-7475, 2020 06 05.
Article En | MEDLINE | ID: mdl-32425046

Intracellular pathogens can thrive within mammalian cells and are inaccessible to many antimicrobial agents. Herein, we present a facile method of enhancing the cell penetrating and antibacterial properties of cationic amphiphilic polyproline helices (CAPHs) with modifications to the hydrophobic moiety at the N-terminus. These altered CAPHs display superior cell penetration within macrophage cells, and in some cases, minimal cytotoxicity. Furthermore, one CAPH, Pentyl-P14 exhibited excellent antibacterial activity against multiple strains of pathogenic bacteria and promoted the clearance of intracellular Shigella within macrophages.


Anti-Bacterial Agents , Peptides , Animals , Anti-Bacterial Agents/pharmacology , Bacteria , Cations , Microbial Sensitivity Tests , Peptides/pharmacology , Protein Structure, Secondary
10.
Sci Rep ; 10(1): 7525, 2020 05 05.
Article En | MEDLINE | ID: mdl-32372011

The limited number of antifungals and the rising frequency of azole-resistant Candida species are growing challenges to human medicine. Drug repurposing signifies an appealing approach to enhance the activity of current antifungal drugs. Here, we evaluated the ability of Pharmakon 1600 drug library to sensitize an azole-resistant Candida albicans to the effect of fluconazole. The primary screen revealed 44 non-antifungal hits were able to act synergistically with fluconazole against the test strain. Of note, 21 compounds, showed aptness for systemic administration and limited toxic effects, were considered as potential fluconazole adjuvants and thus were termed as "repositionable hits". A follow-up analysis revealed pitavastatin displaying the most potent fluconazole chemosensitizing activity against the test strain (ΣFICI 0.05) and thus was further evaluated against 18 isolates of C. albicans (n = 9), C. glabrata (n = 4), and C. auris (n = 5). Pitavastatin displayed broad-spectrum synergistic interactions with both fluconazole and voriconazole against ~89% of the tested strains (ΣFICI 0.05-0.5). Additionally, the pitavastatin-fluconazole combination significantly reduced the biofilm-forming abilities of the tested Candida species by up to 73%, and successfully reduced the fungal burdens in a Caenorhabditis elegans infection model by up to 96%. This study presents pitavastatin as a potent azole chemosensitizing agent that warrant further investigation.


Antifungal Agents/pharmacology , Candida/drug effects , Drug Repositioning/methods , Drug Resistance, Fungal , Quinolines/pharmacology , Voriconazole/pharmacology , Biofilms , Candida albicans/drug effects , Candida glabrata/drug effects , Drug Design , Drug Discovery , Fluconazole/pharmacology , Microbial Sensitivity Tests
11.
Sci Rep ; 10(1): 6089, 2020 04 08.
Article En | MEDLINE | ID: mdl-32269301

Azole antifungals are vital therapeutic options for treating invasive mycotic infections. However, the emergence of azole-resistant isolates combined with limited therapeutic options presents a growing challenge in medical mycology. To address this issue, we utilized microdilution checkerboard assays to evaluate nine stilbene compounds for their ability to interact synergistically with azole drugs, particularly against azole-resistant fungal isolates. Ospemifene displayed the most potent azole chemosensitizing activity, and its combination with itraconazole displayed broad-spectrum synergistic interactions against Candida albicans, Candida auris, Cryptococcus neoformans, and Aspergillus fumigatus (ΣFICI = 0.05-0.50). Additionally, in a Caenorhabditis elegans infection model, the ospemifene-itraconazole combination significantly reduced fungal CFU burdens in infected nematodes by ~75-96%. Nile Red efflux assays and RT-qPCR analysis suggest ospemifene interferes directly with fungal efflux systems, thus permitting entry of azole drugs into fungal cells. This study identifies ospemifene as a novel antifungal adjuvant that augments the antifungal activity of itraconazole against a broad range of fungal pathogens.


ATP-Binding Cassette Transporters/metabolism , Antifungal Agents/pharmacology , Fungal Proteins/metabolism , Itraconazole/pharmacology , Tamoxifen/analogs & derivatives , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/metabolism , Candida/drug effects , Candida/metabolism , Cryptococcus neoformans/drug effects , Cryptococcus neoformans/metabolism , Drug Synergism , Tamoxifen/pharmacology
12.
Sci Rep ; 9(1): 18941, 2019 12 12.
Article En | MEDLINE | ID: mdl-31831822

Candida species are a leading source of healthcare infections globally. The limited number of antifungal drugs combined with the isolation of Candida species, namely C. albicans and C. auris, exhibiting resistance to current antifungals necessitates the development of new therapeutics. The present study tested 85 synthetic phenylthiazole small molecules for antifungal activity against drug-resistant C. albicans. Compound 1 emerged as the most potent molecule, inhibiting growth of C. albicans and C. auris strains at concentrations ranging from 0.25-2 µg/mL. Additionally, compound 1 inhibited growth of other clinically-relevant yeast (Cryptococcus) and molds (Aspergillus) at a concentration as low as 0.50 µg/mL. Compound 1 exhibited rapid fungicidal activity, reducing the burden of C. albicans and C. auris below the limit of detection within 30 minutes. Compound 1 exhibited potent antibiofilm activity, similar to amphotericin B, reducing the metabolic activity of adherent C. albicans and C. auris biofilms by more than 66% and 50%, respectively. Furthermore, compound 1 prolonged survival of Caenorhabditis elegans infected with strains of C. albicans and C. auris, relative to the untreated control. The present study highlights phenylthiazole small molecules, such as compound 1, warrant further investigation as novel antifungal agents for drug-resistant Candida infections.


Antifungal Agents , Biofilms/drug effects , Candida albicans/physiology , Candida/physiology , Thiazoles , Animals , Antifungal Agents/chemical synthesis , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Biofilms/growth & development , Caenorhabditis elegans/microbiology , Chlorocebus aethiops , Thiazoles/chemical synthesis , Thiazoles/chemistry , Thiazoles/pharmacology , Vero Cells
13.
Int J Antimicrob Agents ; 52(6): 754-761, 2018 Dec.
Article En | MEDLINE | ID: mdl-30145250

Candida auris is an emerging multidrug-resistant pathogen implicated in numerous outbreaks worldwide, with a notably high mortality rate (ca. 60%). A significant challenge with treatment of these infections is the resistance of C. auris to most antifungal drugs used clinically. Thus, finding co-drugs capable of overcoming resistance to frontline antifungals is of prime clinical importance. In this study, the ability of the combination of different sulfa drugs with azole antifungals to inhibit the growth of azole-resistant C. auris isolates was evaluated. Among the active sulfa drugs, sulfamethoxazole exhibited the most potent in vitro synergistic interactions with voriconazole and itraconazole. The sulfamethoxazole-voriconazole combination restored voriconazole's fungistatic activity against three of eight voriconazole-resistant clinical isolates. Similarly, the sulfamethoxazole-itraconazole combination restored itraconazole's fungistatic activity against three of four itraconazole-resistant clinical isolates. This activity was further confirmed in vivo in a Caenorhabditis elegans model of C. auris infection. The sulfamethoxazole-voriconazole combination enhanced survival of nematodes infected with C. auris by nearly 70%. Notably, these data indicate that the efficacy of this novel combination is dependent on the underlying mechanism of azole resistance. Mutant strains demonstrating azole resistance by either overproduction of or decreased affinity for the azole target (ERG11p) were found highly to be susceptible to the sulfamethoxazole-azole combination. However, this synergistic interaction was ineffective against mutant strains that demonstrated azole resistance via efflux pump hyperactivity. In conclusion, sulfamethoxazole represents a promising co-drug that can restore the efficacy of certain azole antifungal drugs against some azole-resistant isolates of C. auris.


Antifungal Agents/pharmacology , Candida/drug effects , Drug Synergism , Itraconazole/pharmacology , Sulfamethoxazole/pharmacology , Voriconazole/pharmacology , Animals , Antifungal Agents/therapeutic use , Caenorhabditis elegans , Candida/growth & development , Candidiasis/drug therapy , Disease Models, Animal , Itraconazole/therapeutic use , Sulfamethoxazole/therapeutic use , Survival Analysis , Treatment Outcome , Voriconazole/therapeutic use
14.
ACS Infect Dis ; 4(9): 1300-1305, 2018 09 14.
Article En | MEDLINE | ID: mdl-29979033

A number of pathogenic bacteria reproduce inside mammalian cells and are thus inaccessible to many antimicrobial drugs. Herein, we present a facile method to a focused library of antibacterial agents known as cationic amphiphilic polyproline helices (CAPHs). We identified three CAPHs from the library with superior cell penetration within macrophages and excellent antibacterial action against both Gram-positive and Gram-negative bacteria. These cell-penetrating antibacterial CAPHs have specific subcellular localizations that allow for targeting of pathogenic bacteria at their intracellular niches, a unique feature that promotes the successful clearance of intracellular pathogens ( Salmonella, Shigella, and Listeria) residing within macrophages. Furthermore, the selected CAPHs also significantly reduced bacterial infections in an in vivo model of Caenorhabditis elegans, with minimal in vivo toxicity.


Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Bacterial Infections/microbiology , Peptides/pharmacology , Small Molecule Libraries/pharmacology , Animals , Anti-Bacterial Agents/chemistry , Antimicrobial Cationic Peptides/chemistry , Bacterial Infections/drug therapy , Caenorhabditis elegans , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/growth & development , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/growth & development , Humans , Macrophages/drug effects , Macrophages/microbiology , Peptides/chemistry , Small Molecule Libraries/chemistry
15.
ACS Infect Dis ; 4(3): 403-414, 2018 03 09.
Article En | MEDLINE | ID: mdl-29370698

Globally, invasive fungal infections pose a significant challenge to modern human medicine due to the limited number of antifungal drugs and the rise in resistance to current antifungal agents. A vast majority of invasive fungal infections are caused by species of Candida, Cryptococcus, and Aspergillus. Novel antifungal molecules consisting of unexploited chemical scaffolds with a unique mechanism are a pressing need. The present study identifies a dibromoquinoline compound (4b) with broad-spectrum antifungal activity that inhibits the growth of pertinent species of Candida (chiefly C. albicans), Cryptococcus, and Aspergillus at a concentration of as low as 0.5 µg/mL. Furthermore, 4b, at a subinhibitory concentration, interfered with the expression of two key virulence factors (hyphae and biofilm formation) involved in C. albicans pathogenesis. Three yeast deletion strains ( cox17Δ, ssa1Δ, and aft2Δ) related to metal ion homeostasis were found to be highly sensitive to 4b in growth assays, indicating that the compound exerts its antifungal effect through a unique, previously unexploited mechanism. Supplementing the media with either copper or iron ions reversed the strain sensitivity to 4b, further corroborating that the compound targets metal ion homeostasis. 4b's potent antifungal activity was validated in vivo, as the compound enhanced the survival of Caenorhabditis elegans infected with fluconazole-resistant C. albicans. The present study indicates that 4b warrants further investigation as a novel antifungal agent.


Antifungal Agents/pharmacology , Aspergillus/drug effects , Candida/drug effects , Cryptococcus/drug effects , Ions/metabolism , Metals/metabolism , Quinolines/pharmacology , Animals , Antifungal Agents/chemical synthesis , Antifungal Agents/isolation & purification , Antifungal Agents/therapeutic use , Aspergillus/metabolism , Caenorhabditis elegans/microbiology , Caenorhabditis elegans/physiology , Candida/metabolism , Cryptococcus/metabolism , Culture Media/chemistry , Disease Models, Animal , Homeostasis/drug effects , Mycoses/drug therapy , Quinolines/chemical synthesis , Quinolines/isolation & purification , Quinolines/therapeutic use , Survival Analysis
16.
Article En | MEDLINE | ID: mdl-29263071

Invasive candidiasis presents an emerging global public health challenge due to the emergence of resistance to the frontline treatment options, such as fluconazole. Hence, the identification of other compounds capable of pairing with fluconazole and averting azole resistance would potentially prolong the clinical utility of this important group. In an effort to repurpose drugs in the field of antifungal drug discovery, we explored sulfa antibacterial drugs for the purpose of reversing azole resistance in Candida In this study, we assembled and investigated a library of 21 sulfa antibacterial drugs for their ability to restore fluconazole sensitivity in Candida albicans Surprisingly, the majority of assayed sulfa drugs (15 of 21) were found to exhibit synergistic relationships with fluconazole by checkerboard assay with fractional inhibitory concentration index (ΣFIC) values ranging from <0.0312 to 0.25. Remarkably, five sulfa drugs were able to reverse azole resistance in a clinically achievable range. The structure-activity relationships (SARs) of the amino benzene sulfonamide scaffold as antifungal agents were studied. We also identified the possible mechanism of the synergistic interaction of sulfa antibacterial drugs with azole antifungal drugs. Furthermore, the ability of sulfa antibacterial drugs to inhibit Candida biofilm by 40% in vitro was confirmed. In addition, the effects of sulfa-fluconazole combinations on Candida growth kinetics and efflux machinery were explored. Finally, using a Caenorhabditis elegans infection model, we demonstrated that the sulfa-fluconazole combination does possess potent antifungal activity in vivo, reducing Candida in infected worms by ∼50% compared to the control.


Anti-Bacterial Agents/pharmacology , Antifungal Agents/pharmacology , Azoles/pharmacology , Candida albicans/drug effects , Animals , Biofilms/drug effects , Caenorhabditis elegans/microbiology , Fluconazole/pharmacology , Microbial Sensitivity Tests , Structure-Activity Relationship
17.
Anal Chem ; 89(18): 9822-9829, 2017 09 19.
Article En | MEDLINE | ID: mdl-28813144

Candida albicans is the single most prevalent cause of fungal bloodstream infections worldwide causing significant mortality as high as 50 percent. This high mortality rate is, in part, due to the inability to initiate an effective antifungal therapy early in the disease process. Mortality rates significantly increase after 12 hours of delay in initiating the appropriate antifungal therapy following a positive blood culture. Early administration of appropriate antifungal therapy is hampered by the slow turnovers of the conventional antimicrobial testing techniques, which require days of incubation. To address this unmet need, we explored the potential of employing stimulated Raman scattering (SRS) imaging to probe for metabolic differences between fluconazole-susceptible and -resistant strains at a single cell level in search of a metabolic signature. Metabolism is integral to pathogenicity. Since only a few hours are needed to observe a full metabolic cycle in C. albicans, metabolic profiling provides an avenue for rapid antimicrobial susceptibility testing. C-H frequency (2850 cm-1) SRS imaging revealed a substantial difference in lipogenesis between the fluconazole-susceptible and -resistant C. albicans. Exposure to fluconazole, an antimicrobial drug that targets ergosterol biosynthesis, only affected the lipogenesis in the susceptible strain. These results show that single cell metabolic imaging via SRS microscopy can be used for rapid detection of antimicrobial susceptibility.


Antifungal Agents/pharmacology , Azoles/pharmacology , Candida albicans/drug effects , Drug Resistance, Fungal/drug effects , Lipogenesis , Spectrum Analysis, Raman/methods , Antifungal Agents/chemistry , Azoles/chemistry , Biomarkers/analysis , Biomarkers/metabolism , Candida albicans/cytology , Cells, Cultured , Microbial Sensitivity Tests , Optical Imaging , Spectrum Analysis, Raman/instrumentation
18.
Chem Commun (Camb) ; 53(53): 7238-7241, 2017 Jun 29.
Article En | MEDLINE | ID: mdl-28492641

A novel and efficient palladium-catalyzed aminocarbonylative lactonization of amino propargylic alcohols has been developed to provide rapid access to various bicyclic lactones especially dihydropyrrole-fused furanones, which are novel structures and have not been explored in biological and medicinal settings. This method can also be used to access ß-lactone products such as 16. Preliminary biological evaluations revealed that compounds 13h and 13s demonstrated promising activity against Clostridium difficile and compounds 13h, 13k, 13s, and 16b showed activity against several important fungal pathogens.

19.
Biochim Biophys Acta Gen Subj ; 1861(1 Pt A): 3002-3010, 2017 Jan.
Article En | MEDLINE | ID: mdl-27712973

BACKGROUND: Ebselen, an organoselenium compound and a clinically safe molecule has been reported to possess potent antifungal activity, but its antifungal mechanism of action and in vivo antifungal activity remain unclear. METHODS: The antifungal effect of ebselen was tested against Candida albicans, C. glabrata, C. tropicalis, C. parapsilosis, Cryptococcus neoformans, and C. gattii clinical isolates. Chemogenomic profiling and biochemical assays were employed to identify the antifungal target of ebselen. Ebselen's antifungal activity in vivo was investigated in a Caenorhabditis elegans animal model. RESULTS: Ebselen exhibits potent antifungal activity against both Candida spp. and Cryptococcus spp., at concentrations ranging from 0.5 to 2µg/ml. Ebselen rapidly eradicates a high fungal inoculum within 2h of treatment. Investigation of the drug's antifungal mechanism of action indicates that ebselen depletes intracellular glutathione (GSH) levels, leading to increased production of reactive oxygen species (ROS), and thereby disturbs the redox homeostasis in fungal cells. Examination of ebselen's in vivo antifungal activity in two Caenorhabditis elegans models of infection demonstrate that ebselen is superior to conventional antifungal drugs (fluconazole, flucytosine and amphotericin) in reducing Candida and Cryptococcus fungal load. CONCLUSION: Ebselen possesses potent antifungal activity against clinically relevant isolates of both Candida and Cryptococcus by regulating GSH and ROS production. The potent in vivo antifungal activity of ebselen supports further investigation for repurposing it for use as an antifungal agent. GENERAL SIGNIFICANCE: The present study shows that ebselen targets glutathione and also support that glutathione as a potential target for antifungal drug development.


Antifungal Agents/pharmacology , Azoles/pharmacology , Candida/cytology , Cryptococcus/cytology , Glutathione/biosynthesis , Organoselenium Compounds/pharmacology , Reactive Oxygen Species/metabolism , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/microbiology , Candida/drug effects , Candida/growth & development , Cryptococcus/drug effects , Cryptococcus/growth & development , Glutathione/pharmacology , Isoindoles , Kinetics , Microbial Sensitivity Tests , Microbial Viability/drug effects
...