Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Infect Dis ; 227(7): 901-906, 2023 04 12.
Article in English | MEDLINE | ID: mdl-36611269

ABSTRACT

Influenza-associated pulmonary aspergillosis (IAPA) is a feared complication in patients with influenza tracheobronchitis, especially those receiving corticosteroids. Herein, we established a novel IAPA mouse model with low-inoculum Aspergillus infection and compared outcomes in mice with and without cortisone acetate (CA) immunosuppression. CA was an independent predictor of increased morbidity/mortality in mice with IAPA. Early antifungal treatment with liposomal amphotericin B was pivotal to improve IAPA outcomes in CA-immunosuppressed mice, even after prior antiviral therapy with oseltamivir. In summary, our model recapitulates key clinical features of IAPA and provides a robust preclinical platform to study the pathogenesis and treatment of IAPA.


Subject(s)
Aspergillosis , Influenza, Human , Pulmonary Aspergillosis , Animals , Mice , Humans , Influenza, Human/complications , Influenza, Human/drug therapy , Pulmonary Aspergillosis/complications , Pulmonary Aspergillosis/drug therapy , Antifungal Agents/therapeutic use , Aspergillosis/drug therapy , Adrenal Cortex Hormones/therapeutic use , Aspergillus fumigatus
2.
Med Mycol ; 59(1): 102-105, 2021 Jan 04.
Article in English | MEDLINE | ID: mdl-32678869

ABSTRACT

Vasopressors are frequently given in hemodynamically unstable patients with severe Candida sepsis. While catecholamines can aggravate sepsis-induced immune dysfunction and modulate bacterial virulence traits, their impact on fungal pathogenicity is poorly understood. Using IncuCyte time-lapse microscopy and a fruit fly candidiasis model, we studied growth rates, morphogenesis, stress tolerance, and virulence of C. albicans cocultured with epinephrine and norepinephrine. We found that pharmacologically attainable catecholamine serum concentrations caused minimal changes to in vitro growth kinetics, filamentation, and fungal resistance to thermal or oxidative stress. Similarly, exposure of C. albicans to catecholamines did not alter the survival of infected flies.


Subject(s)
Candida albicans/drug effects , Candida albicans/pathogenicity , Candidiasis/drug therapy , Epinephrine/blood , Epinephrine/pharmacology , Norepinephrine/blood , Norepinephrine/pharmacology , Virulence/drug effects , Growth/drug effects , Humans , Morphogenesis/drug effects , Stress, Physiological/drug effects
3.
J Infect Dis ; 222(6): 989-994, 2020 08 17.
Article in English | MEDLINE | ID: mdl-32432714

ABSTRACT

Pharmacological immune checkpoint blockade has revolutionized oncological therapies, and its remarkable success has sparked interest in expanding checkpoint inhibitor therapy in infectious diseases. Herein, we evaluated the efficacy of programmed cell death protein 1 (PD-1) blockade in a murine invasive pulmonary aspergillosis model. We found that, compared with isotype-treated infected control mice, anti-PD-1-treated mice had improved survival, reduced fungal burden, increased lung concentrations of proinflammatory cytokines and neutrophil-attracting chemokines, and enhanced pulmonary leukocyte accumulation. Furthermore, combined treatment with anti-PD-1 and caspofungin resulted in a significant survival benefit compared with caspofungin or anti-PD-1 therapy alone, indicating a synergistic effect between PD-1 inhibitors and immunomodulatory antifungal agents.


Subject(s)
Antifungal Agents/pharmacology , Aspergillus/drug effects , Caspofungin/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Invasive Pulmonary Aspergillosis/metabolism , Invasive Pulmonary Aspergillosis/microbiology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Immunohistochemistry , Invasive Pulmonary Aspergillosis/drug therapy , Mice , Microbial Sensitivity Tests , Programmed Cell Death 1 Receptor/metabolism
4.
Article in English | MEDLINE | ID: mdl-30455245

ABSTRACT

Breakthrough mucormycosis in patients receiving isavuconazole prophylaxis or therapy has been reported. We compared the impact of isavuconazole and voriconazole exposure on the virulence of clinical isolates of Aspergillus fumigatus and different Mucorales species in a Drosophila melanogaster infection model. In contrast to A. fumigatus, a hypervirulent phenotype was found in all tested Mucorales upon preexposure to either voriconazole or isavuconazole. These findings may contribute to the explanation of breakthrough mucormycosis in isavuconazole-treated patients.


Subject(s)
Antifungal Agents/pharmacology , Aspergillus fumigatus/pathogenicity , Mucorales/pathogenicity , Nitriles/pharmacology , Pyridines/pharmacology , Triazoles/pharmacology , Animals , Aspergillus fumigatus/drug effects , Drosophila melanogaster , Female , Mucorales/drug effects , Rhizopus/drug effects , Rhizopus/pathogenicity , Virulence
5.
J Antimicrob Chemother ; 74(7): 1904-1910, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31225606

ABSTRACT

OBJECTIVES: Candida auris is an emerging, often MDR, yeast pathogen. Efficient animal models are needed to study its pathogenicity and treatment. Therefore, we developed a C. auris fruit fly infection model. METHODS: TollI-RXA/Tollr632 female flies were infected with 10 different C. auris strains from the CDC Antimicrobial Resistance bank panel. We used three clinical Candida albicans strains as controls. For drug protection assays, fly survival was assessed along with measurement of fungal burden (cfu/g tissue) and histopathology in C. auris-infected flies fed with fluconazole- or posaconazole-containing food. RESULTS: Despite slower in vitro growth, all 10 C. auris isolates caused significantly greater mortality than C. albicans in infected flies, with >80% of C. auris-infected flies dying by day 7 post-infection (versus 67% with C. albicans, P < 0.001-0.005). Comparison of C. auris isolates from different geographical clades revealed more rapid in vitro growth of South American isolates and greater virulence in infected flies, whereas the aggregative capacity of C. auris strains had minimal impact on their growth and pathogenicity. Survival protection and decreased fungal burden of fluconazole- or posaconazole-fed flies infected with two C. auris strains were in line with the isolates' disparate in vitro azole susceptibility. High reproducibility of survival curves for both non-treated and antifungal-treated infected flies was seen, with coefficients of variation of 0.00-0.31 for 7 day mortality. CONCLUSIONS: Toll-deficient flies could provide a fast, reliable and inexpensive model to study pathogenesis and drug activity in C. auris candidiasis.


Subject(s)
Antifungal Agents/pharmacology , Azoles/pharmacology , Candida/drug effects , Candida/pathogenicity , Candidiasis/drug therapy , Candidiasis/microbiology , Animals , Animals, Genetically Modified , Biopsy , Candida albicans/drug effects , Candida albicans/pathogenicity , Candidiasis/pathology , Disease Models, Animal , Drosophila melanogaster , Microbial Sensitivity Tests , Virulence
6.
J Mater Sci Mater Med ; 29(5): 70, 2018 May 11.
Article in English | MEDLINE | ID: mdl-29752591

ABSTRACT

While antibiotic-eluting polymethylmethacrylate space maintainers have shown efficacy in the treatment of bacterial periprosthetic joint infection and osteomyelitis, antifungal-eluting space maintainers are associated with greater limitations for treatment of fungal musculoskeletal infections including limited elution concentration and duration. In this study, we have designed a porous econazole-eluting space maintainer capable of greater inhibition of fungal growth than traditional solid space maintainers. The eluted econazole demonstrated bioactivity in a concentration-dependent manner against the most common species responsible for fungal periprosthetic joint infection as well as staphylococci. Lastly, these porous space maintainers retain compressive mechanical properties appropriate to maintain space before definitive repair of the joint or bony defect.


Subject(s)
Antifungal Agents/chemistry , Biocompatible Materials , Econazole/chemistry , Mycoses/drug therapy , Prosthesis-Related Infections/drug therapy , Antifungal Agents/pharmacology , Aspergillus fumigatus/drug effects , Candida albicans/drug effects , Econazole/pharmacology , Materials Testing , Polymethyl Methacrylate , Porosity , Staphylococcus aureus/drug effects
7.
Clin Infect Dis ; 65(2): 216-225, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28379304

ABSTRACT

BACKGROUND: Azole-resistant aspergillosis in high-risk patients with hematological malignancy or hematopoietic stem cell transplantation (HSCT) is a cause of concern. METHODS: We examined changes over time in triazole minimum inhibitory concentrations (MICs) of 290 sequential Aspergillus isolates recovered from respiratory sources during 1999-2002 (before introduction of the Aspergillus-potent triazoles voriconazole and posaconazole) and 2003-2015 at MD Anderson Cancer Center. We also tested for polymorphisms in ergosterol biosynthetic genes (cyp51A, erg3C, erg1) in the 37 Aspergillus fumigatus isolates isolated from both periods that had non-wild-type (WT) MICs. For the 107 patients with hematologic cancer and/or HSCT with invasive pulmonary aspergillosis, we correlated in vitro susceptibility with 42-day mortality. RESULTS: Non-WT MICs were found in 37 (13%) isolates and was only low level (MIC <8 mg/L) in all isolates. Higher-triazole MICs were more frequent in the second period and were Aspergillus-species specific, and only encountered in A. fumigatus. No polymorphisms in cyp51A, erg3C, erg1 genes were identified. There was no correlation between in vitro MICs with 42-day mortality in patients with invasive pulmonary aspergillosis, irrespective of antifungal treatment. Asian race (odds ratio [OR], 20.9; 95% confidence interval [CI], 2.5-173.5; P = .005) and azole exposure in the prior 3 months (OR, 9.6; 95% CI, 1.9-48.5; P = .006) were associated with azole resistance. CONCLUSIONS: Non-WT azole MICs in Aspergillus are increasing and this is associated with prior azole exposure in patients with hematologic cancer or HSCT. However, no correlation of MIC with outcome of aspergillosis was found in our patient cohort.


Subject(s)
Antifungal Agents/pharmacology , Aspergillus/drug effects , Invasive Pulmonary Aspergillosis/microbiology , Tertiary Healthcare , Triazoles/pharmacology , Adult , Antifungal Agents/therapeutic use , Aspergillosis/drug therapy , Aspergillosis/microbiology , Aspergillus/genetics , Aspergillus/isolation & purification , Aspergillus fumigatus/drug effects , Cohort Studies , Cytochrome P-450 Enzyme System/genetics , Drug Resistance, Fungal/genetics , Ergosterol/biosynthesis , Female , Fungal Proteins/genetics , Hematologic Neoplasms/complications , Hematologic Neoplasms/microbiology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Invasive Pulmonary Aspergillosis/drug therapy , Invasive Pulmonary Aspergillosis/mortality , Male , Microbial Sensitivity Tests , Polymorphism, Genetic , Prospective Studies , Treatment Outcome , Triazoles/therapeutic use , Voriconazole/pharmacology , Voriconazole/therapeutic use , Young Adult
8.
Article in English | MEDLINE | ID: mdl-27993850

ABSTRACT

Systemic candidiasis is a leading cause of nosocomial bloodstream infection with a high mortality rate despite treatment. Immune-based strategies are needed to improve outcomes. We previously reported that genetic deficiency in the chemokine receptor CCR1 improves survival and ameliorates tissue damage in Candida-infected mice. Here, we found that treatment of immunocompetent Candida-infected mice with the CCR1-selective antagonist BL5923 improves survival, decreases the kidney fungal burden, and protects from renal tissue injury.


Subject(s)
Acute Kidney Injury/prevention & control , Candida albicans/drug effects , Candidiasis, Invasive/drug therapy , Hemorrhage/prevention & control , Piperazines/pharmacology , Receptors, CCR1/antagonists & inhibitors , Acute Kidney Injury/immunology , Acute Kidney Injury/microbiology , Acute Kidney Injury/mortality , Animals , Candida albicans/growth & development , Candida albicans/pathogenicity , Candidiasis, Invasive/immunology , Candidiasis, Invasive/microbiology , Candidiasis, Invasive/mortality , Drug Administration Schedule , Female , Gene Expression , Hemorrhage/immunology , Hemorrhage/microbiology , Hemorrhage/mortality , Host-Pathogen Interactions , Mice , Mice, Inbred C57BL , Receptors, CCR1/genetics , Receptors, CCR1/immunology , Survival Analysis
9.
Proc Natl Acad Sci U S A ; 111(29): 10660-5, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-25002471

ABSTRACT

Clinical-grade T cells are genetically modified ex vivo to express chimeric antigen receptors (CARs) to redirect their specificity to target tumor-associated antigens in vivo. We now have developed this molecular strategy to render cytotoxic T cells specific for fungi. We adapted the pattern-recognition receptor Dectin-1 to activate T cells via chimeric CD28 and CD3-ζ (designated "D-CAR") upon binding with carbohydrate in the cell wall of Aspergillus germlings. T cells genetically modified with the Sleeping Beauty system to express D-CAR stably were propagated selectively on artificial activating and propagating cells using an approach similar to that approved by the Food and Drug Administration for manufacturing CD19-specific CAR(+) T cells for clinical trials. The D-CAR(+) T cells exhibited specificity for ß-glucan which led to damage and inhibition of hyphal growth of Aspergillus in vitro and in vivo. Treatment of D-CAR(+) T cells with steroids did not compromise antifungal activity significantly. These data support the targeting of carbohydrate antigens by CAR(+) T cells and provide a clinically appealing strategy to enhance immunity for opportunistic fungal infections using T-cell gene therapy.


Subject(s)
Aspergillosis/immunology , Aspergillosis/therapy , Bioengineering/methods , Carbohydrates/antagonists & inhibitors , Opportunistic Infections/immunology , Opportunistic Infections/therapy , T-Lymphocytes/immunology , Animals , Antigens, CD19/metabolism , Aspergillosis/microbiology , Aspergillosis/pathology , Aspergillus/drug effects , Aspergillus/physiology , Dexamethasone/pharmacology , Humans , Hyphae/drug effects , Hyphae/physiology , Immunophenotyping , Lectins, C-Type/metabolism , Lymphocyte Activation/drug effects , Mice , Opportunistic Infections/pathology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/drug effects
10.
J Infect Dis ; 214(1): 114-21, 2016 07 01.
Article in English | MEDLINE | ID: mdl-26984141

ABSTRACT

BACKGROUND: Mucormycosis is a destructive invasive mold infection afflicting patients with diabetes and hematologic malignancies. Patients with diabetes are often treated with statins, which have been shown to have antifungal properties. We sought to examine the effects of statins on Rhizopus oryzae, a common cause of mucormycosis. METHODS: Clinical strains of R. oryzae were exposed to lovastatin, atorvastatin, and simvastatin and the minimum inhibitory concentrations (MICs) were determined. R. oryzae germination, DNA fragmentation, susceptibility to oxidative stress, and ability to damage endothelial cells were assessed. We further investigated the impact of exposure to lovastatin on the virulence of R. oryzae RESULTS: All statins had MICs of >64 µg/mL against R. oryzae Exposure of R. oryzae to statins decreased germling formation, induced DNA fragmentation, and attenuated damage to endothelial cells independently of the expression of GRP78 and CotH. Additionally, R. oryzae exposed to lovastatin showed macroscopic loss of melanin, yielded increased susceptibility to the oxidative agent peroxide, and had attenuated virulence in both fly and mouse models of mucormycosis. CONCLUSIONS: Exposure of R. oryzae to statins at concentrations below their MICs decreased virulence both in vitro and in vivo. Further investigation is warranted into the use of statins as adjunctive therapy in mucormycosis.


Subject(s)
Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Mucormycosis/drug therapy , Rhizopus/drug effects , Virulence/drug effects , Animals , Atorvastatin/pharmacology , Atorvastatin/therapeutic use , Diptera/drug effects , Endoplasmic Reticulum Chaperone BiP , Female , Humans , Lovastatin/pharmacology , Lovastatin/therapeutic use , Mice , Microbial Sensitivity Tests , Simvastatin/pharmacology , Simvastatin/therapeutic use , Spores, Fungal/drug effects , Texas
11.
Antimicrob Agents Chemother ; 59(12): 7830-2, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26392499

ABSTRACT

Triazole prophylaxis has become the norm in patients with hematological malignancies. Breakthrough infections caused by Mucorales during triazole prophylaxis remain a challenging problem. We found that preexposure of Rhizopus oryzae to antifungal triazoles (fluconazole, voriconazole, posaconazole, and itraconazole) did not modify the in vitro susceptibility of Rhizopus oryzae to posaconazole. In contrast, preexposure of Rhizopus to triazoles was associated with the enhanced in vitro susceptibility of R. oryzae to amphotericin B. Preexposure to posaconazole did not alter the virulence of R. oryzae in the fly model of mucormycosis.


Subject(s)
Antifungal Agents/pharmacology , Fluconazole/pharmacology , Itraconazole/pharmacology , Rhizopus/drug effects , Triazoles/pharmacology , Voriconazole/pharmacology , Agar , Animals , Culture Media/chemistry , Disease Models, Animal , Drosophila melanogaster/drug effects , Drosophila melanogaster/microbiology , Female , Microbial Sensitivity Tests , Mucormycosis/drug therapy , Mucormycosis/microbiology , Mucormycosis/mortality , Rhizopus/growth & development , Rhizopus/pathogenicity , Survival Analysis , Virulence
12.
J Infect Dis ; 210(9): 1471-5, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-24837401

ABSTRACT

High concentrations of methylprednisolone (0.32 mg/mL) accelerated growth and attenuated spontaneous apoptosis of Exserohilum rostratum in vitro. Injection of E. rostratum conidia preexposed to 0.32 mg/mL of methylprednisolone for 7 days in immunocompetent flies led to increased mortality and a higher fungal burden. Exposure to methylprednisolone could enhance the virulence of E. rostratum.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Ascomycota/drug effects , Methylprednisolone/pharmacology , Mycoses/drug therapy , Animals , Ascomycota/growth & development , Ascomycota/physiology , Drosophila melanogaster/drug effects , Drosophila melanogaster/microbiology , Humans , In Vitro Techniques , Mycoses/microbiology
13.
Antimicrob Agents Chemother ; 58(11): 6767-72, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25182639

ABSTRACT

We used two established neutropenic murine models of pulmonary aspergillosis and mucormycosis to explore the association between the posaconazole area under the concentration-time curve (AUC)-to-MIC ratio (AUC/MIC) and treatment outcome. Posaconazole serum pharmacokinetics were verified in infected mice to ensure that the studied doses reflected human exposures with the oral suspension, delayed-release tablet, and intravenous formulations of posaconazole. Sinopulmonary infections were then induced in groups of neutropenic mice with Aspergillus fumigatus strain 293 (posaconazole MIC, 0.5 mg/liter) or Rhizopus oryzae strain 969 (posaconazole MIC, 2 mg/liter) and treated with escalating daily dosages of oral posaconazole, which was designed to achieve AUCs ranging from 1.10 to 392 mg · h/liter. After 5 days of treatment, lung fungal burden was analyzed by quantitative real-time PCR. The relationships of the total drug AUC/MIC and the treatment response were similar in both models, with 90% effective concentrations (EC90s) corresponding to an AUC/MIC threshold of 76 (95% confidence interval [CI], 46 to 102) for strain 293 versus 87 (95% CI, 66 to 101) for strain 969. Using a provisional AUC/MIC target of >100, these exposures correlated with minimum serum posaconazole concentrations (Cmins) of 1.25 mg/liter for strain 293 and 4.0 mg/liter for strain 969. The addition of deferasirox, but not liposomal amphotericin or caspofungin, improved the activity of a suboptimal posaconazole regimen (AUC/MIC, 33) in animals with pulmonary mucormycosis. However, no combination was as effective as the high-dose posaconazole monotherapy regimen (AUC/MIC, 184). Our analysis suggests that posaconazole pharmacodynamics are similar for A. fumigatus and R. oryzae when indexed to pathogen MICs.


Subject(s)
Antifungal Agents/pharmacokinetics , Invasive Pulmonary Aspergillosis/drug therapy , Mucormycosis/drug therapy , Triazoles/pharmacokinetics , Triazoles/therapeutic use , Amphotericin B/pharmacology , Animals , Antifungal Agents/therapeutic use , Area Under Curve , Aspergillosis/drug therapy , Aspergillosis/microbiology , Aspergillus fumigatus/drug effects , Benzoates/pharmacology , Caspofungin , Deferasirox , Disease Models, Animal , Drug Therapy, Combination , Echinocandins/pharmacology , Female , Invasive Pulmonary Aspergillosis/microbiology , Lipopeptides , Lung/microbiology , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Neutropenia , Rhizopus/drug effects , Treatment Outcome , Triazoles/pharmacology
14.
J Infect Dis ; 207(7): 1066-74, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23303813

ABSTRACT

In invasive pulmonary aspergillosis, direct invasion and occlusion of pulmonary vasculature by Aspergillus hyphae causes tissue hypoxia, which is enhanced by secreted fungal metabolites that downregulate compensatory angiogenic signaling pathways. We assessed the effects of basic fibroblast growth factor (bFGF) and vascular endothelial growth factor (VEGF) on survival rates, fungal burden, and in situ angiogenesis in a murine invasive pulmonary aspergillosis model. bFGF and VEGF monotherapy significantly increased survival rates and potentiated the activity of amphotericin B. bFGF-containing regimens were associated with reduced tissue fungal burdens. bFGF and VEGF reversed the antiangiogenic activity of Aspergillus fumigatus; however, VEGF induced the formation of immature neovessels, providing an explanation for its lesser efficacy. Treatment with bFGF plus amphotericin B was associated with neutrophil influx into Aspergillus-infected pulmonary tissue, suggesting that this combination limits fungal growth through neutrophil trafficking. Vasculogenic pathways are unexplored targets for the treatment of invasive pulmonary aspergillosis and may potentiate both innate immunity and antifungal drug activity against A. fumigatus.


Subject(s)
Angiogenesis Inducing Agents/therapeutic use , Antifungal Agents/therapeutic use , Aspergillosis/drug therapy , Aspergillus fumigatus/pathogenicity , Fibroblast Growth Factor 2/therapeutic use , Neovascularization, Physiologic/drug effects , Vascular Endothelial Growth Factor A/therapeutic use , Amphotericin B/therapeutic use , Animals , Aspergillosis/microbiology , Aspergillosis/pathology , Aspergillus fumigatus/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Therapy, Combination , Female , Humans , Immunohistochemistry , Lung/microbiology , Lung/pathology , Lung Diseases, Fungal/drug therapy , Lung Diseases, Fungal/microbiology , Lung Diseases, Fungal/pathology , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Neutrophils/drug effects , Recombinant Proteins/therapeutic use , Survival Analysis
15.
J Infect Dis ; 208(1): 83-91, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23547139

ABSTRACT

BACKGROUND: Mini-host models are simple experimental systems to study host-pathogen interactions. We adapted a Drosophila melanogaster infection model to evaluate the in vivo effect of different mechanisms of linezolid (LNZ) resistance in Staphylococcus aureus. METHODS: Fly survival was evaluated after infection with LNZ-resistant S. aureus strains NRS119 (which has mutations in 23S ribosomal RNA [rRNA]), CM-05 and 004-737X (which carry cfr), LNZ-susceptible derivatives of CM-05 and 004-737X (which lack cfr), and ATCC 29213 (an LNZ-susceptible control). Flies were then fed food mixed with LNZ (concentration, 15-500 µg/mL). Results were compared to those in mouse peritonitis, using LNZ via oral gavage at 80 and 120 mg/kg every 12 hours. RESULTS: LNZ at 500 µg/mL in fly food protected against all strains, while concentrations of 15-250 µg/mL failed to protect against NRS119 (survival, 1.6%-20%). An in vivo effect of cfr was only detected at concentrations of 30 and 15 µg/mL. In the mouse peritonitis model, LNZ (at doses that mimic human pharmacokinetics) protected mice from challenge with the cfr+ 004-737X strain but was ineffective against the NRS119 strain, which carried 23S rRNA mutations. CONCLUSIONS: The fly model offers promising advantages to dissect the in vivo effect of LNZ resistance in S. aureus, and findings from this model appear to be concordant with those from the mouse peritonitis model.


Subject(s)
Acetamides/pharmacology , Anti-Bacterial Agents/pharmacology , Oxazolidinones/pharmacology , Staphylococcal Infections/drug therapy , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Drosophila melanogaster/drug effects , Drosophila melanogaster/microbiology , Drug Resistance, Bacterial , Female , Linezolid , Mice , Mice, Inbred ICR , Staphylococcus aureus/drug effects
16.
mBio ; 15(4): e0341323, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38415653

ABSTRACT

Invasive aspergillosis (IA) is a common and deadly mold infection in immunocompromised patients. As morbidity and mortality of IA are primarily driven by poor immune defense, adjunct immunotherapies, such as chimeric antigen receptor (CAR) T cells, are direly needed. Here, we propose a novel approach to generate Aspergillus fumigatus (AF)-CAR T cells using the single-chain variable fragment domain of monoclonal antibody AF-269-5 and a lentiviral vector system. These cells successfully targeted mature hyphal filaments of representative clinical and reference AF isolates and elicited a potent release of cytotoxic effectors and type 1 T cell cytokines. Furthermore, AF-CAR T cells generated from peripheral blood mononuclear cells of four healthy human donors and expanded with either of three cytokine stimulation regimens (IL-2, IL-2 + IL-21, or IL-7 + IL-15) significantly suppressed mycelial growth of AF-293 after 18 hours of co-culture and synergized with the immunomodulatory antifungal agent caspofungin to control hyphal growth for 36 hours. Moreover, cyclophosphamide-immunosuppressed NSG mice with invasive pulmonary aspergillosis that received two doses of 5 million AF-CAR T cells (6 and 48 hours after AF infection) showed significantly reduced morbidity on day 4 post-infection (P < 0.001) and significantly improved 7-day survival (P = 0.049) compared with mice receiving non-targeting control T cells, even without concomitant antifungal chemotherapy. In conclusion, we developed a novel lentiviral strategy to obtain AF-CAR T cells with high targeting efficacy, yielding significant anti-AF activity in vitro and short-term protection in vivo. Our approach could serve as an important steppingstone for future clinical translation of antifungal CAR T-cell therapy after further refinement and thorough preclinical evaluation.IMPORTANCEInvasive aspergillosis (IA) remains a formidable cause of morbidity and mortality in patients with hematologic malignancies and those undergoing hematopoietic stem cell transplantation. Despite the introduction of several new Aspergillus-active antifungals over the last 30 years, the persisting high mortality of IA in the setting of continuous and profound immunosuppression is a painful reminder of the major unmet need of effective antifungal immune enhancement therapies. The success of chimeric antigen receptor (CAR) T-cell therapy in cancer medicine has inspired researchers to translate this approach to opportunistic infections, including IA. Aiming to refine anti-Aspergillus CAR T-cell therapy and improve its feasibility for future clinical translation, we herein developed and validated a novel antibody-based CAR construct and lentiviral transduction method to accelerate the production of CAR T cells with high targeting efficacy against Aspergillus fumigatus. Our unique approach could provide a promising platform for future clinical translation of CAR T-cell-based antifungal immunotherapy.


Subject(s)
Aspergillosis , Receptors, Chimeric Antigen , Humans , Mice , Animals , Aspergillus fumigatus/genetics , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/therapeutic use , Interleukin-2 , Antifungal Agents/therapeutic use , Lentivirus/genetics , Leukocytes, Mononuclear , Aspergillosis/drug therapy , Aspergillus , T-Lymphocytes , Cytokines
17.
Antimicrob Agents Chemother ; 57(9): 4444-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23836166

ABSTRACT

We studied the effect of noninvasive radiofrequency-induced hyperthermia on the viability of Aspergillus fumigatus hyphae in vitro. Radiofrequency-induced hyperthermia resulted in significant (>70%, P < 0.0001) hyphal damage in a time and thermal dose-dependent fashion as assessed by XTT [(sodium 2,3,-bis(2-methoxy-4-nitro-5-sulfophenyl)-5-[(phenylamino)-carbonyl] (1)-2H-tetrazolium inner salt)], DiBAC [bis-(1,3-dibutylbarbituric acid) trimethine oxonol] staining, and transmission electron microscopy. For comparison, water bath hyperthermia was used over the range of 45 to 55°C to study hyphal damage. Radiofrequency-induced hyperthermia resulted in severe damage to the outer fibrillar layer of hyphae at a shorter treatment time compared to water bath hyperthermia. Our preliminary data suggest that radiofrequency-induced hyperthermia might be an additional therapeutic approach to use in the management of mold infections.


Subject(s)
Aspergillus fumigatus/ultrastructure , Hyphae/ultrastructure , Radio Waves , Aspergillus fumigatus/growth & development , Barbiturates , Fluorescent Dyes , Hot Temperature , Hyphae/growth & development , Isoxazoles , Microbial Viability , Tetrazolium Salts
18.
Microb Pathog ; 55: 16-20, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23232438

ABSTRACT

BACKGROUND: USA300 is a uniquely successful methicillin-resistant Staphylococcus aureus (MRSA) clone that has been associated with Panton-Valentine leukocidin (PVL) production and severe infections. However, conflicting experimental and epidemiological data exist regarding the virulence of USA300 relative to other MRSA clones. We aimed to address this issue using Drosophila melanogaster as a model host to study strain and PVL-dependent variations in virulence among MRSA clinical isolates. RESULTS: We studied the relative virulence of 39 MRSA isolates: 17 (43%) were PFGE type USA300. Lethal MRSA infection was reproducibly induced both in wild-type (WT) and Toll-deficient D. melanogaster. USA300 strains had significantly lower lethality than non-USA300 strains in a WT background but not in Toll-deficient flies. PFGE type (USA300 versus non-USA300) and PVL status did not affect the response to treatment with linezolid. Virulence was similar in strains with high vancomycin MIC (≥2 µg/mL) versus those with vancomycin MIC<2 µg/mL. CONCLUSIONS: D. melanogaster is a potentially useful model host to study pathogenicity and response to antibiotic treatment in S. aureus. Our results imply that the attenuated virulence of PVL(+)/USA300 requires intact host innate immunity.


Subject(s)
Acetamides/administration & dosage , Anti-Bacterial Agents/administration & dosage , Drosophila melanogaster , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Oxazolidinones/administration & dosage , Staphylococcal Infections/drug therapy , Animals , Disease Models, Animal , Drosophila melanogaster/microbiology , Humans , Linezolid , Methicillin-Resistant Staphylococcus aureus/physiology , Staphylococcal Infections/microbiology , Virulence/drug effects
19.
Methods Mol Biol ; 2517: 299-316, 2022.
Article in English | MEDLINE | ID: mdl-35674964

ABSTRACT

While mammalian models remain the gold standard to study invasive mycoses, mini-host invertebrate models have provided complementary platforms for explorative investigations of fungal pathogenesis, host-pathogen interplay, and antifungal therapy. Specifically, our group has established Toll-deficient Drosophila melanogaster flies as a facile and cost-effective model organism to study candidiasis, and we have recently expanded these studies to the emerging and frequently multidrug-resistant yeast pathogen Candida auris. Our proof-of-concept data suggest that fruit flies could hold a great promise for large-scale applications in antifungal drug discovery and the screening of C. auris (mutant) libraries with disparate pathogenic capacity. This chapter discusses the advantages and limitations of D. melanogaster to study C. auris candidiasis and provides a step-by-step guide for establishing and troubleshooting C. auris infection and antifungal treatment of Toll-deficient flies along with essential downstream readouts.


Subject(s)
Candidiasis , Drosophila melanogaster , Animals , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Candida , Candida auris , Candidiasis/drug therapy , Candidiasis/microbiology , Drosophila melanogaster/microbiology , Mammals , Microbial Sensitivity Tests , Saccharomyces cerevisiae
20.
mSphere ; 7(2): e0081721, 2022 04 27.
Article in English | MEDLINE | ID: mdl-35224979

ABSTRACT

Candida auris is a globally spreading yeast pathogen causing bloodstream infections with high mortality in critically ill patients. The inherent antifungal drug resistance of most C. auris isolates and threat of multidrug-resistant strains create a need for adjunct immunotherapeutic strategies. While C. albicans candidemia was shown to induce immune paralysis and activation of inhibitory immune checkpoints, in vivo data on host responses to C. auris bloodstream infection are lacking as is an immunocompetent murine infection model to study the immunopathology and immunotherapy of C. auris sepsis. Therefore, herein, we developed an immunocompetent C. auris sepsis model by intravenously infecting C57BL/6 mice with 1.5 × 108 to 8 × 108 yeast cells of aggregate-forming (AR-0384) and nonaggregative (AR-0381) C. auris reference isolates. Both isolates caused reproducible, inoculum-dependent increasing morbidity, mortality, and fungal burden in kidney tissue. Notably, morbidity and mortality outcomes were partially decoupled from fungal burden, suggesting a role of additional modulators of disease severity such as host immune responses. Flow cytometric analyses of splenic immune cells revealed significant upregulation of the programmed cell death protein 1 (PD-1) on T cells and its ligand PD-L1 on macrophages from mice infected with C. auris AR-0384 compared to uninfected mice. PD-L1 expression on macrophages from AR-0384-infected mice strongly correlated with fungal tissue burden (Spearman's rank correlation coefficient [ρ] = 0.95). Altogether, our findings suggest that C. auris sepsis promotes a suppressive immune phenotype through PD-1/PD-L1 induction, supporting further exploration of PD-1/PD-L1 blockade as an immunotherapeutic strategy to mitigate C. auris candidiasis. IMPORTANCE Health authorities consider Candida auris to be one of the most serious emerging nosocomial pathogens due to its transmissibility, resistance to disinfection procedures, and frequent antifungal drug resistance. The frequency of multidrug-resistant C. auris isolates necessitates the development of novel therapeutic platforms, including immunotherapy. However, in vivo data on host interactions with C. auris are scarce, compounded by the lack of reliable immunocompetent mammalian models of C. auris candidemia. Herein, we describe a C. auris sepsis model in immunocompetent C57BL/6 mice and demonstrate reproducible and inoculum-dependent acute infection with both aggregate-forming and nonaggregative reference isolates from different clades. Furthermore, we show that C. auris sepsis induces upregulation of the PD-1/PD-L1 immune checkpoint pathway in infected mice, raising the potential of a therapeutic benefit of immune checkpoint blockade. Our immunocompetent model of C. auris sepsis could provide a facile preclinical platform to thoroughly investigate immune checkpoint blockade and combination therapy with antifungals.


Subject(s)
Candidemia , Candidiasis, Invasive , Immune Checkpoint Proteins , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/therapeutic use , Candida/genetics , Candida albicans , Candida auris , Candidemia/drug therapy , Disease Models, Animal , Humans , Mammals , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/therapeutic use , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL