Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 491
Filtrar
1.
J Mater Chem B ; 12(34): 8357-8365, 2024 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-39101217

RESUMO

Skin infections are a major threat to human health. Early diagnosis of bacterial infections is of great significance for implementing protective measures on the skin. Therefore, in this study, we designed an electrospun membrane (PPBT) for visual monitoring of colonized bacteria and responsive antibacterial ability. Specifically, the acidity of the microenvironment caused by bacterial metabolism was applied to drive the color change of bromothymol blue (BTB) on the PPBT membrane from green to yellow, thereby facilitating the early warning of infection and timely treatment. Within 4 h, different concentrations of Staphylococcus aureus (∼105 CFU mL-1), Escherichia coli (∼105 CFU mL-1), Pseudomonas aeruginosa (∼105 CFU mL-1) and Candida albicans (∼104 CFU mL-1) were visually monitored. Moreover, as the local acidity was enhanced via microbial metabolism, ZIF-8 nanoparticles loaded with TCS (TCS@ZIF-8) on the PPBT membrane could release TCS in an acid-responsive manner. At the same time, ROS were generated under 405 nm irradiation to achieve synergistic antibacterial ability. Experiments confirmed that the PPBT membrane has ideal and controllable antibacterial features based on acid responsive release and a synergistic photocatalytic antibacterial mechanism after monitoring. Therefore, the PPBT membrane developed in this work provides a feasible solution for bacterial monitoring and inactivation devices. More importantly, it can be beneficial for meeting the needs of clinical diagnosis and timely treatment of bacterial infection.


Assuntos
Membranas Artificiais , Elétrons , Staphylococcus aureus , Candida albicans , Pseudomonas aeruginosa/metabolismo , Escherichia coli/metabolismo , Infecções Estafilocócicas/metabolismo , Candidíase/metabolismo , Infecções por Pseudomonas , Infecções por Escherichia coli , Estruturas Metalorgânicas/química , Estruturas Metalorgânicas/farmacologia , Imidazóis/química , Imidazóis/farmacologia , Antibacterianos/química , Antibacterianos/farmacologia , Trifosfato de Adenosina/metabolismo , Sistemas de Liberação de Medicamentos , Nanopartículas Metálicas/química , Nanofibras/química
2.
Sci Signal ; 17(849): eads1861, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39137247
3.
PLoS Pathog ; 20(7): e1012389, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39078851

RESUMO

Ergosterol is essential for fungal cell membrane integrity and growth, and numerous antifungal drugs target ergosterol. Inactivation or modification of ergosterol biosynthetic genes can lead to changes in antifungal drug susceptibility, filamentation and stress response. Here, we found that the ergosterol biosynthesis gene ERG251 is a hotspot for point mutations during adaptation to antifungal drug stress within two distinct genetic backgrounds of Candida albicans. Heterozygous point mutations led to single allele dysfunction of ERG251 and resulted in azole tolerance in both genetic backgrounds. This is the first known example of point mutations causing azole tolerance in C. albicans. Importantly, single allele dysfunction of ERG251 in combination with recurrent chromosome aneuploidies resulted in bona fide azole resistance. Homozygous deletions of ERG251 caused increased fitness in low concentrations of fluconazole and decreased fitness in rich medium, especially at low initial cell density. Homozygous deletions of ERG251 resulted in accumulation of ergosterol intermediates consistent with the fitness defect in rich medium. Dysfunction of ERG251, together with FLC exposure, resulted in decreased accumulation of the toxic sterol (14-ɑ-methylergosta-8,24(28)-dien-3ß,6α-diol) and increased accumulation of non-toxic alternative sterols. The altered sterol composition of the ERG251 mutants had pleiotropic effects on transcription, filamentation, and stress responses including cell membrane, osmotic and oxidative stress. Interestingly, while dysfunction of ERG251 resulted in azole tolerance, it also led to transcriptional upregulation of ZRT2, a membrane-bound Zinc transporter, in the presence of FLC, and overexpression of ZRT2 is sufficient to increase azole tolerance in wild-type C. albicans. Finally, in a murine model of systemic infection, homozygous deletion of ERG251 resulted in decreased virulence while the heterozygous deletion mutants maintain their pathogenicity. Overall, this study demonstrates that single allele dysfunction of ERG251 is a recurrent and effective mechanism of acquired azole tolerance. We propose that altered sterol composition resulting from ERG251 dysfunction mediates azole tolerance as well as pleiotropic effects on stress response, filamentation and virulence.


Assuntos
Antifúngicos , Candida albicans , Candidíase , Farmacorresistência Fúngica , Ergosterol , Proteínas Fúngicas , Candida albicans/efeitos dos fármacos , Candida albicans/genética , Candida albicans/metabolismo , Antifúngicos/farmacologia , Camundongos , Farmacorresistência Fúngica/genética , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/genética , Animais , Candidíase/microbiologia , Candidíase/metabolismo , Candidíase/tratamento farmacológico , Ergosterol/metabolismo , Azóis/farmacologia , Esteróis/metabolismo , Fenótipo , Estresse Fisiológico , Testes de Sensibilidade Microbiana , Fluconazol/farmacologia
4.
Nat Commun ; 15(1): 5746, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38982056

RESUMO

Candida albicans and Staphylococcus aureus are two commonly associated pathogens that cause nosocomial infections with high morbidity and mortality. Our prior and current work using a murine model of polymicrobial intra-abdominal infection (IAI) demonstrates that synergistic lethality is driven by Candida-induced upregulation of functional S. aureus α-toxin leading to polymicrobial sepsis and organ damage. In order to determine the candidal effector(s) mediating enhanced virulence, an unbiased screen of C. albicans transcription factor mutants was undertaken revealing that zcf13Δ/Δ fails to drive augmented α-toxin or lethal synergism during co-infection. A combination of transcriptional and phenotypic profiling approaches shows that ZCF13 regulates genes involved in pentose metabolism, including RBK1 and HGT7 that contribute to fungal ribose catabolism and uptake, respectively. Subsequent experiments reveal that ribose inhibits the staphylococcal agr quorum sensing system and concomitantly represses toxicity. Unlike wild-type C. albicans, zcf13Δ/Δ did not effectively utilize ribose during co-culture or co-infection leading to exogenous ribose accumulation and agr repression. Forced expression of RBK1 and HGT7 in the zcf13Δ/Δ mutant fully restores pathogenicity during co-infection. Collectively, our results detail the interwoven complexities of cross-kingdom interactions and highlight how intermicrobial metabolism impacts polymicrobial disease pathogenesis with devastating consequences for the host.


Assuntos
Candida albicans , Candidíase , Coinfecção , Infecções Intra-Abdominais , Infecções Estafilocócicas , Staphylococcus aureus , Camundongos , Candida albicans/fisiologia , Staphylococcus aureus/fisiologia , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/patologia , Candidíase/metabolismo , Candidíase/patologia , Coinfecção/metabolismo , Coinfecção/patologia , Toxoide Estafilocócico/metabolismo , Proteínas Fúngicas/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Bactérias/metabolismo , Transativadores/metabolismo , Percepção de Quorum , Infecções Intra-Abdominais/metabolismo , Infecções Intra-Abdominais/microbiologia , Infecções Intra-Abdominais/patologia , Açúcares/metabolismo , Ribose/metabolismo , Modelos Animais de Doenças
5.
Invest Ophthalmol Vis Sci ; 65(6): 13, 2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38848078

RESUMO

Purpose: Fungal keratitis (FK) is an invasive corneal infection associated with significant risk to vision. Although the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) signaling pathway has been recognized for its role in defending against viral infections, its involvement in FK still remains largely unclear. This study sought to elucidate the contribution of the cGAS/STING signaling pathway to the pathogenesis of FK. Methods: The expression of cGAS/STING signaling components was assessed in a murine model of Candida albicans keratitis through RNA sequencing, western blot analysis, immunofluorescence staining, and real-time PCR. Both genetic (utilizing Sting1gt/gt mice) and pharmacological (using C176) interventions were employed to inhibit STING activity, allowing for the evaluation of resultant pathogenic alterations in FK using slit-lamp examination, clinical scoring, hematoxylin and eosin (H&E) staining, fungal culture, and RNA sequencing. Subconjunctival administration of the NOD-like receptor protein 3 (NLRP3) inflammasome inhibitor MCC950 was performed to evaluate FK manifestations following STING activity blockade. Furthermore, the impact of the STING agonist diABZI on FK progression was investigated. Results: Compared to uninfected corneas, those infected with C. albicans exhibited increased expression of cGAS/STING signaling components, as well as its elevated activity. Inhibiting cGAS/STING signaling exacerbated the advancement of FK, as evidenced by elevated clinical scores, augmented fungal load, and heightened inflammatory response, including NLRP3 inflammasome activation and pyroptosis. Pharmacological inhibition of the NLRP3 inflammasome effectively mitigated the exacerbated FK by suppressing STING activity. Conversely, pre-activation of STING exacerbated FK progression compared to the PBS control, characterized by increased fungal burden and reinforced inflammatory infiltration. Conclusions: This study demonstrates the essential role of the cGAS/STING signaling pathway in FK pathogenesis and highlights the necessity of its proper activation for the host against FK.


Assuntos
Candida albicans , Candidíase , Modelos Animais de Doenças , Infecções Oculares Fúngicas , Proteínas de Membrana , Nucleotidiltransferases , Transdução de Sinais , Animais , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/genética , Infecções Oculares Fúngicas/microbiologia , Infecções Oculares Fúngicas/metabolismo , Camundongos , Candida albicans/fisiologia , Candidíase/microbiologia , Candidíase/metabolismo , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real , Ceratite/microbiologia , Ceratite/metabolismo , Western Blotting , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Feminino , Úlcera da Córnea/microbiologia , Úlcera da Córnea/metabolismo , Inflamassomos/metabolismo
6.
Cell Mol Life Sci ; 81(1): 280, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38918243

RESUMO

Candida albicans is among the most prevalent invasive fungal pathogens for immunocompromised individuals and novel therapeutic approaches that involve immune response modulation are imperative. Absent in melanoma 2 (AIM2), a pattern recognition receptor for DNA sensing, is well recognized for its involvement in inflammasome formation and its crucial role in safeguarding the host against various pathogenic infections. However, the role of AIM2 in host defense against C. albicans infection remains uncertain. This study reveals that the gene expression of AIM2 is induced in human and mouse innate immune cells or tissues after C. albicans infection. Furthermore, compared to their wild-type (WT) counterparts, Aim2-/- mice surprisingly exhibit resistance to C. albicans infection, along with reduced inflammation in the kidneys post-infection. The resistance of Aim2-/- mice to C. albicans infection is not reliant on inflammasome or type I interferon production. Instead, Aim2-/- mice display lower levels of apoptosis in kidney tissues following infection than WT mice. The deficiency of AIM2 in macrophages, but not in dendritic cells, results in a phenocopy of the resistance observed in Aim2-/- mice against C. albican infection. The treatment of Clodronate Liposome, a reagent that depletes macrophages, also shows the critical role of macrophages in host defense against C. albican infection in Aim2-/- mice. Furthermore, the reduction in apoptosis is observed in Aim2-/- mouse macrophages following infection or treatment of DNA from C. albicans in comparison with controls. Additionally, higher levels of AKT activation are observed in Aim2-/- mice, and treatment with an AKT inhibitor reverses the host resistance to C. albicans infection. The findings collectively demonstrate that AIM2 exerts a negative regulatory effect on AKT activation and enhances macrophage apoptosis, ultimately compromising host defense against C. albicans infection. This suggests that AIM2 and AKT may represent promising therapeutic targets for the management of fungal infections.


Assuntos
Apoptose , Candida albicans , Candidíase , Proteínas de Ligação a DNA , Macrófagos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Animais , Macrófagos/metabolismo , Macrófagos/imunologia , Macrófagos/microbiologia , Candidíase/imunologia , Candidíase/microbiologia , Candidíase/metabolismo , Candidíase/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Camundongos , Humanos , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Inflamassomos/metabolismo , Imunidade Inata , Rim/patologia , Rim/metabolismo , Rim/microbiologia
7.
Sci Rep ; 14(1): 14723, 2024 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-38926392

RESUMO

Invasive candidiasis (IC) is a notable healthcare-associated fungal infection, characterized by high morbidity, mortality, and substantial treatment costs. Candida albicans emerges as a principal pathogen in this context. Recent academic advancements have shed light on the critical role of exosomes in key biological processes, such as immune responses and antigen presentation. This burgeoning body of research underscores the potential of exosomes in the realm of medical diagnostics and therapeutics, particularly in relation to fungal infections like IC. The exploration of exosomal functions in the pathophysiology of IC not only enhances our understanding of the disease but also opens new avenues for innovative therapeutic interventions. In this investigation, we focus on exosomes (Exos) secreted by macrophages, both uninfected and those infected with C. albicans. Our objective is to extract and analyze these exosomes, delving into the nuances of their protein compositions and subgroups. To achieve this, we employ an innovative technique known as Proximity Barcoding Assay (PBA). This methodology is pivotal in our quest to identify novel biological targets, which could significantly enhance the diagnostic and therapeutic approaches for C. albicans infection. The comparative analysis of exosomal contents from these two distinct cellular states promises to yield insightful data, potentially leading to breakthroughs in understanding and treating this invasive fungal infection. In our study, we analyzed differentially expressed proteins in exosomes from macrophages and C. albicans -infected macrophages, focusing on proteins such as ACE2, CD36, CAV1, LAMP2, CD27, and MPO. We also examined exosome subpopulations, finding a dominant expression of MPO in the most prevalent subgroup, and a distinct expression of CD36 in cluster14. These findings are crucial for understanding the host response to C. albicans and may inform targeted diagnostic and therapeutic approaches. Our study leads us to infer that MPO and CD36 proteins may play roles in the immune escape mechanisms of C. albicans. Additionally, the CD36 exosome subpopulations, identified through our analysis, could serve as potential biomarkers and therapeutic targets for C. albicans infection. This insight opens new avenues for understanding the infection's pathology and developing targeted treatments.


Assuntos
Biomarcadores , Antígenos CD36 , Candida albicans , Candidíase , Exossomos , Macrófagos , Exossomos/metabolismo , Biomarcadores/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiologia , Macrófagos/imunologia , Antígenos CD36/metabolismo , Candidíase/diagnóstico , Candidíase/microbiologia , Candidíase/metabolismo , Candidíase/imunologia , Humanos , Animais , Camundongos
8.
Free Radic Biol Med ; 222: 344-360, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38945457

RESUMO

Innovative solutions for rapid protection against broad-spectrum infections are very important in dealing with complex infection environments. We utilized a functionally inactive mutated endolysin protein of Streptococcus pneumoniae (ΔA146Ply) to immunize mice against pneumonic infections by multidrug-resistant bacteria, Candida albicans and influenza virus type A. ΔA146Ply protection relied on both immunized tissue-resident and monocyte-derived alveolar macrophages and inhibited infection induced ferroptosis that upregulated expression of GPX4 (glutathione peroxidase) in alveolar macrophages. Ferroptosis resistance endowed macrophages with enhanced phagocytosis by inhibiting lipid peroxidation during infection. Moreover, we demonstrated ΔA146Ply upregulated GPX4 through the TLR4/IRG1/NRF2 pathway. ΔA146Ply also induced ferroptosis inhibition and phagocytosis improvement in human monocytes. This mode of action is a novel and potentially prophylactic and rapid broad-spectrum anti-infection mechanism. Our study provides new insights into protective interventions that act by regulating ferroptosis to improve multiple pathogen resistance via GPX4 targeting.


Assuntos
Ferroptose , Macrófagos Alveolares , Fator 2 Relacionado a NF-E2 , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Streptococcus pneumoniae , Receptor 4 Toll-Like , Animais , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/genética , Camundongos , Streptococcus pneumoniae/patogenicidade , Ferroptose/genética , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/genética , Receptor 4 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Humanos , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/microbiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Fagocitose , Candida albicans/patogenicidade , Candidíase/imunologia , Candidíase/genética , Candidíase/microbiologia , Candidíase/metabolismo , Regulação para Cima , Feminino
9.
Cell Host Microbe ; 32(6): 964-979.e7, 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38754418

RESUMO

The gut microbiota is closely linked to atherosclerosis. However, the role of intestinal fungi, essential members of the complex microbial community, in atherosclerosis is poorly understood. Herein, we show that gut fungi dysbiosis is implicated in patients with dyslipidemia, characterized by higher levels of Candida albicans (C. albicans), which are positively correlated with plasma total cholesterol and low-density lipoprotein-cholesterol (LDL-C) levels. Furthermore, C. albicans colonization aggravates atherosclerosis progression in a mouse model of the disease. Through gain- and loss-of-function studies, we show that an intestinal hypoxia-inducible factor 2α (HIF-2α)-ceramide pathway mediates the effect of C. albicans. Mechanistically, formyl-methionine, a metabolite of C. albicans, activates intestinal HIF-2α signaling, which drives increased ceramide synthesis to accelerate atherosclerosis. Administration of the HIF-2α selective antagonist PT2385 alleviates atherosclerosis in mice by reducing ceramide levels. Our findings identify a role for intestinal fungi in atherosclerosis progression and highlight the intestinal HIF-2α-ceramide pathway as a target for atherosclerosis treatment.


Assuntos
Aterosclerose , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Candida albicans , Ceramidas , Transdução de Sinais , Animais , Candida albicans/metabolismo , Aterosclerose/microbiologia , Aterosclerose/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Camundongos , Humanos , Ceramidas/metabolismo , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Masculino , Microbioma Gastrointestinal/fisiologia , Intestinos/microbiologia , Intestinos/patologia , Disbiose/microbiologia , Feminino , Candidíase/microbiologia , Candidíase/metabolismo
10.
Open Biol ; 14(5): 230315, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38806144

RESUMO

Candida glabrata is an important pathogen causing invasive infection associated with a high mortality rate. One mechanism that causes the failure of Candida eradication is an increase in regulatory T cells (Treg), which play a major role in immune suppression and promoting Candida pathogenicity. To date, how C. glabrata induces a Treg response remains unclear. Dendritic cells (DCs) recognition of fungi provides the fundamental signal determining the fate of the T-cell response. This study investigated the interplay between C. glabrata and DCs and its effect on Treg induction. We found that C. glabrata ß-glucan was a major component that interacted with DCs and consequently mediated the Treg response. Blocking the binding of C. glabrata ß-glucan to dectin-1 and complement receptor 3 (CR3) showed that CR3 activation in DCs was crucial for the induction of Treg. Furthermore, a ligand-receptor binding assay showed the preferential binding of C. glabrata ß-glucan to CR3. Our data suggest that C. glabrata ß-glucan potentially mediates the Treg response, probably through CR3-dependent activation in DCs. This study contributes new insights into immune modulation by C. glabrata that may lead to a better design of novel immunotherapeutic strategies for invasive C. glabrata infection.


Assuntos
Candida glabrata , Células Dendríticas , Antígeno de Macrófago 1 , Linfócitos T Reguladores , beta-Glucanas , Candida glabrata/metabolismo , Candida glabrata/patogenicidade , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , beta-Glucanas/metabolismo , beta-Glucanas/farmacologia , Animais , Antígeno de Macrófago 1/metabolismo , Camundongos , Lectinas Tipo C/metabolismo , Candidíase/imunologia , Candidíase/microbiologia , Candidíase/metabolismo , Camundongos Endogâmicos C57BL
11.
PLoS Pathog ; 20(5): e1012225, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38739655

RESUMO

Biofilm formation by the fungal pathogen Candida albicans is the basis for its ability to infect medical devices. The metabolic gene ERG251 has been identified as a target of biofilm transcriptional regulator Efg1, and here we report that ERG251 is required for biofilm formation but not conventional free-living planktonic growth. An erg251Δ/Δ mutation impairs biofilm formation in vitro and in an in vivo catheter infection model. In both in vitro and in vivo biofilm contexts, cell number is reduced and hyphal length is limited. To determine whether the mutant defect is in growth or some other aspect of biofilm development, we examined planktonic cell features in a biofilm-like environment, which was approximated with sealed unshaken cultures. Under those conditions, the erg251Δ/Δ mutation causes defects in growth and hyphal extension. Overexpression in the erg251Δ/Δ mutant of the paralog ERG25, which is normally expressed more weakly than ERG251, partially improves biofilm formation and biofilm hyphal content, as well as growth and hyphal extension in a biofilm-like environment. GC-MS analysis shows that the erg251Δ/Δ mutation causes a defect in ergosterol accumulation when cells are cultivated under biofilm-like conditions, but not under conventional planktonic conditions. Overexpression of ERG25 in the erg251Δ/Δ mutant causes some increase in ergosterol levels. Finally, the hypersensitivity of efg1Δ/Δ mutants to the ergosterol inhibitor fluconazole is reversed by ERG251 overexpression, arguing that reduced ERG251 expression contributes to this efg1Δ/Δ phenotype. Our results indicate that ERG251 is required for biofilm formation because its high expression levels are necessary for ergosterol synthesis in a biofilm-like environment.


Assuntos
Biofilmes , Candida albicans , Candidíase , Proteínas Fúngicas , Biofilmes/crescimento & desenvolvimento , Candida albicans/metabolismo , Candida albicans/genética , Candida albicans/fisiologia , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/genética , Animais , Candidíase/microbiologia , Candidíase/metabolismo , Hifas/metabolismo , Camundongos , Regulação Fúngica da Expressão Gênica , Ergosterol/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Mutação
12.
Nat Microbiol ; 9(1): 95-107, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38168615

RESUMO

The host type I interferon (IFN) pathway is a major signature of inflammation induced by the human fungal pathogen, Candida albicans. However, the molecular mechanism for activating this pathway in the host defence against C. albicans remains unknown. Here we reveal that mice lacking cyclic GMP-AMP synthase (cGAS)-stimulator of IFN genes (STING) pathway components had improved survival following an intravenous challenge by C. albicans. Biofilm-associated C. albicans DNA packaged in extracellular vesicles triggers the cGAS-STING pathway as determined by induction of interferon-stimulated genes, IFNß production, and phosphorylation of IFN regulatory factor 3 and TANK-binding kinase 1. Extracellular vesicle-induced activation of type I IFNs was independent of the Dectin-1/Card9 pathway and did not require toll-like receptor 9. Single nucleotide polymorphisms in cGAS and STING potently altered inflammatory cytokine production in human monocytes challenged by C. albicans. These studies provide insights into the early innate immune response induced by a clinically significant fungal pathogen.


Assuntos
Candidíase , Interferon Tipo I , Animais , Camundongos , Candida albicans/patogenicidade , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Imunidade Inata , Interferon Tipo I/metabolismo , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Transdução de Sinais , Candidíase/metabolismo , Candidíase/patologia
13.
PLoS Pathog ; 20(1): e1011902, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38166150

RESUMO

Fungal infections have emerged as a major concern among immunocompromised patients, causing approximately 2 million deaths each year worldwide. However, the regulatory mechanisms underlying antifungal immunity remain elusive and require further investigation. The E3 ligase Trim26 belongs to the tripartite motif (Trim) protein family, which is involved in various biological processes, including cell proliferation, antiviral innate immunity, and inflammatory responses. Herein, we report that Trim26 exerts protective antifungal immune functions after fungal infection. Trim26-deficient mice are more susceptible to fungemia than their wild-type counterparts. Mechanistically, Trim26 restricts inflammatory neutrophils infiltration and limits proinflammatory cytokine production, which can attenuate kidney fungal load and renal damage during Candida infection. Trim26-deficient neutrophils showed higher proinflammatory cytokine expression and impaired fungicidal activity. We further demonstrated that excessive neutrophils infiltration in the kidney was because of the increased production of chemokines CXCL1 and CXCL2, which are mainly synthesized in the macrophages or dendritic cells of Trim26-deficient mice after Candida albicans infections. Together, our study findings unraveled the vital role of Trim26 in regulating antifungal immunity through the regulation of inflammatory neutrophils infiltration and proinflammatory cytokine and chemokine expression during candidiasis.


Assuntos
Candidíase , Neutrófilos , Animais , Camundongos , Antifúngicos , Candida albicans , Candidíase/metabolismo , Candidíase/microbiologia , Citocinas/metabolismo , Infiltração de Neutrófilos , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases
14.
Microbes Infect ; 26(4): 105305, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38296157

RESUMO

The liver, and more specifically, the liver sinusoidal endothelial cells, constitute the beginning of one of the most important responses for the elimination of hematogenously disseminated Candida albicans. Therefore, we aimed to study the mechanisms involved in the interaction between these cells and C. albicans. Transcriptomics-based analysis showed an increase in the expression of genes related to the immune response (including receptors, cytokines, and adhesion molecules), as well as to aerobic glycolysis. Further in vitro analyses showed that IL-6 production in response to C. albicans is controlled by MyD88- and SYK-pathways, suggesting an involvement of Toll-like and C-type lectin receptors and the subsequent activation of the MAP-kinases and c-Fos/AP-1 transcription factor. In addition, liver sinusoidal endothelial cells undergo metabolic reprogramming towards aerobic glycolysis induced by C. albicans, as confirmed by the increased Extracellular Acidification Rate and the overexpression of enolase (Eno2), hexonikase (Hk2) and glucose transporter 1 (Slc2a1). In conclusion, these results indicate that the hepatic endothelium responds to C. albicans by increasing aerobic glycolysis and promoting an inflammatory environment.


Assuntos
Candida albicans , Células Endoteliais , Glicólise , Fígado , Candida albicans/imunologia , Células Endoteliais/metabolismo , Células Endoteliais/microbiologia , Animais , Fígado/metabolismo , Fígado/microbiologia , Quinase Syk/metabolismo , Interleucina-6/metabolismo , Interleucina-6/genética , Camundongos , Fator 88 de Diferenciação Mieloide/metabolismo , Inflamação/metabolismo , Perfilação da Expressão Gênica , Candidíase/imunologia , Candidíase/microbiologia , Candidíase/metabolismo
15.
Int J Mol Sci ; 24(14)2023 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-37511508

RESUMO

Endothelial and epithelial cells are morphologically different and play a critical role in host defense during Candida albicans infection. Both cells respond to C. albicans infection by activating various signaling pathways and gene expression patterns. Their interactions with these pathogens can have beneficial and detrimental effects, and a better understanding of these interactions can help guide the development of new therapies for C. albicans infection. To identify the differences and similarities between human endothelial and oral epithelial cell transcriptomics during C. albicans infection, we performed consensus WGCNA on 32 RNA-seq samples by relating the consensus modules to endothelial-specific modules and analyzing the genes connected. This analysis resulted in the identification of 14 distinct modules. We demonstrated that the magenta module correlates significantly with C. albicans infection in each dataset. In addition, we found that the blue and cyan modules in the two datasets had opposite correlation coefficients with a C. albicans infection. However, the correlation coefficients and p-values between the two datasets were slightly different. Functional analyses of the hub of genes from endothelial cells elucidated the enrichment in TNF, AGE-RAGE, MAPK, and NF-κB signaling. On the other hand, glycolysis, pyruvate metabolism, amino acid, fructose, mannose, and vitamin B6 metabolism were enriched in epithelial cells. However, mitophagy, necroptosis, apoptotic processes, and hypoxia were enriched in both endothelial and epithelial cells. Protein-protein interaction analysis using STRING and CytoHubba revealed STAT3, SNRPE, BIRC2, and NFKB2 as endothelial hub genes, while RRS1, SURF6, HK2, and LDHA genes were identified in epithelial cells. Understanding these similarities and differences may provide new insights into the pathogenesis of C. albicans infections and the development of new therapeutic targets and interventional strategies.


Assuntos
Candida albicans , Candidíase , Humanos , Candida albicans/genética , Redes Reguladoras de Genes , Células Endoteliais/metabolismo , Consenso , Candidíase/metabolismo , Células Epiteliais/metabolismo , Proteínas Nucleares/genética
16.
Int J Clin Pract ; 2022: 5279323, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35832797

RESUMO

Histatins (Hsts) are considered a prominent member of antimicrobial peptides rich in histidine, bearing antifungal activity against Candida species. Hst5 is the most effective among them. Although Hst5 is not found in the cervicovaginal fluid, it has been detected in the human serum. Saliva acts as a mirror, reflecting the cause and effect relationship between several diseases. We aimed to show the salivary Hst5 levels with vaginal candidiasis. Women in the reproductive age group (18-50 years) were enrolled in the study. Patients and controls were classified based on the presence or absence of vaginal discharge suggestive of candidiasis, respectively. Vaginal and salivary samples were collected from all the women. Vaginal samples were cultured for the growth of Candida species. Salivary samples were tested by protein electrophoresis to detect Hst5 levels, and the results were compared between the two groups. A total of 80 women were included in this study. The mean age of women in vaginal candidiasis and control groups was 34.25 ± 8.06 and 36.83 ± 7.29 years, respectively. Candida species were isolated from the vaginal samples of the patient group (34 C. albicans, 6 non-Candida albicans) but not from the control group. Hst5 levels in the patient and control group were found to be 0.0571 ± 0.003 ng/mL and 0.0641 ± 0,0031 ng/mL, respectively. Hst5 levels were found to be significantly lower in the vaginal candidiasis group (p=0.001). We conclude that decreased salivary Hst5 levels in women are associated with vaginal candidiasis. Candida infection is a cause or result of lower salivary Hst5 levels, and it may be an important finding for the etiopathogenesis, diagnosis, and treatment of the disease, but further analysis is needed.


Assuntos
Candidíase , Histatinas , Adolescente , Adulto , Antifúngicos/uso terapêutico , Candida , Candidíase/tratamento farmacológico , Candidíase/metabolismo , Feminino , Histatinas/metabolismo , Humanos , Pessoa de Meia-Idade , Saliva/metabolismo , Adulto Jovem
17.
Zhonghua Yi Xue Za Zhi ; 102(25): 1924-1930, 2022 Jul 05.
Artigo em Chinês | MEDLINE | ID: mdl-35768392

RESUMO

Objective: To investigate the immune mechanism of human airway epithelial cell injury induced by invasion of Candida albicans with different biofilm formation abilities. Methods: Twenty-five strains of Candida albicans isolated and cultured in General Hospital of Ningxia Medical University from June to December 2019 were selected, and quality control strain SC5314 was used as the standard strain. An in vitro model of Candida albicans biofilm was established, and the biofilm formation ability of different Candida albicans was detected by crystal violet staining and enzyme plate method. The absorbance value at 570 nm (A570) was determined by enzyme plate method. A570≥0.5, 0.250.05). Conclusion: Strong biofilm Candida albican can inhibit cell proliferation, disrupt the integrity of epithelial cells and induce cell damage by down-regulating the expression of cell proliferation-related protein.


Assuntos
Candida albicans , Candidíase , Antifúngicos , Candida albicans/fisiologia , Candidíase/metabolismo , Células Epiteliais/metabolismo , Fluconazol/metabolismo , Fluconazol/farmacologia , Fator Estimulador de Colônias de Granulócitos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos
18.
Carbohydr Polym ; 283: 119178, 2022 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-35153023

RESUMO

The aim of this study was to combine fluconazole (FZ)-loaded solid lipid nanoparticles (FZ-SLNs) and chitosan films (C-films) for the potential administration of FZ across the buccal mucosa using a Box-Behnken design. The chitosan films containing FZ-SLNs (C-FS-films) and C-films were prepared using a film casting method. The ATR-FTIR analysis confirmed the presence of hydrogen bonds between the NH3+ groups of chitosan and the OH or COO- groups of glyceryl monostearate in the films. Additionally, FESEM analysis of the morphology of C-FS-films revealed the presence of FZ-SLNs in the films. Permeation studies using porcine buccal mucosa demonstrated that FZ from the C-FS-films was more permeable than in C-films. The antifungal activity of the C-FS-films was evaluated against Candida albicans, and inhibition zones were observed. Thus, C-FS-films represent an exciting drug carrier for the treatment of candidiasis via the buccal mucosa.


Assuntos
Antifúngicos/farmacologia , Candidíase/tratamento farmacológico , Quitosana/química , Fluconazol/farmacologia , Lipossomos/química , Nanopartículas/química , Adesividade , Administração Bucal , Animais , Candida albicans/efeitos dos fármacos , Candidíase/metabolismo , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos/métodos , Glicerídeos/química , Mucosa Bucal/metabolismo , Tamanho da Partícula , Espectroscopia de Infravermelho com Transformada de Fourier/métodos , Suínos
19.
J Microbiol ; 60(4): 402-410, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35157222

RESUMO

Acute lung injury caused by Candida albicans could result in high mortality and morbidity. MicroRNA-155 (miR-155) and suppressor of cytokine signaling 1 (SOCS1) have been believed to play a key in the regulation of inflammatory response. Whether miR-155/SOCS1 axis could regulate the acute lung injury caused by C. albicans has not been reported. The acute lung injury animal model was established with acute infection of C. albicans. miR-155 inhibitor, miR-155 mimic, and sh-SOCS1 were constructed. The binding site between miR-155 and SOCS1 was identified with dual luciferase reporter assay. Knockdown of miR-155 markedly inhibited the germ tube formation of C. albicans. Knockdown of miR-155 significantly up-regulated the expression of SOCS1, and the binding site between miR-155 and SOCS1 was identified. Knockdown of miR-155 improved the acute lung injury, suppressed inflammatory factors and fungus loading through SOCS1. Knockdown of SOCS1 greatly reversed the influence of miR-155 inhibitor on the cell apoptosis in vitro. The improvement of acute lung injury caused by C. albicans, suppression of inflammatory response and C. albicans infection, and inhibitor of cell apoptosis were achieved by knocking down miR-155 through SOCS1. This research might provide a new thought for the prevention and treatment of acute lung injury caused by C. albicans through targeting miR-155/SOCS1 axis.


Assuntos
Lesão Pulmonar Aguda , Candida albicans , Candidíase , MicroRNAs , Proteína 1 Supressora da Sinalização de Citocina , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/metabolismo , Animais , Candida albicans/genética , Candida albicans/metabolismo , Candidíase/genética , Candidíase/metabolismo , Candidíase/microbiologia , Regulação para Baixo , Inflamação/metabolismo , Inflamação/microbiologia , MicroRNAs/genética , MicroRNAs/metabolismo , Proteína 1 Supressora da Sinalização de Citocina/genética , Proteína 1 Supressora da Sinalização de Citocina/metabolismo
20.
Gut Microbes ; 14(1): 2004798, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35086419

RESUMO

Candida albicans (C. albicans) is an opportunistic pathogen causing infections ranging from superficial to life-threatening disseminated infections. In a susceptible host, C. albicans is able to translocate through the gut barrier, promoting its dissemination into deeper organs. C. albicans hyphae can invade human epithelial cells by two well-documented mechanisms: epithelial-driven endocytosis and C. albicans-driven active penetration. One mechanism by which host cells protect themselves against intracellular C. albicans is termed autophagy. The protective role of autophagy during C. albicans infection has been investigated in myeloid cells; however, far less is known regarding the role of this process during the infection of epithelial cells. In the present study, we investigated the role of autophagy-related proteins during the infection of epithelial cells, including intestinal epithelial cells and gut explants, by C. albicans. Using cell imaging, we show that key molecular players of the autophagy machinery (LC3-II, PI3P, ATG16L1, and WIPI2) were recruited at Candida invasion sites. We deepened these observations by electron microscopy analyses that reveal the presence of autophagosomes in the vicinity of invading hyphae. Importantly, these events occur during active penetration of C. albicans into host cells and are associated with plasma membrane damage. In this context, we show that the autophagy-related key proteins ATG5 and ATG16L1 contribute to plasma membrane repair mediated by lysosomal exocytosis and participate in protecting epithelial cells against C. albicans-induced cell death. Our findings provide a novel mechanism by which epithelial cells, forming the first line of defense against C. albicans in the gut, can react to limit C. albicans invasion.


Assuntos
Autofagia , Candida albicans/fisiologia , Candidíase/microbiologia , Membrana Celular/microbiologia , Células Epiteliais/microbiologia , Proteína 5 Relacionada à Autofagia/genética , Proteína 5 Relacionada à Autofagia/metabolismo , Proteínas Relacionadas à Autofagia/genética , Proteínas Relacionadas à Autofagia/metabolismo , Candida albicans/genética , Candidíase/genética , Candidíase/metabolismo , Candidíase/fisiopatologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Microbioma Gastrointestinal , Interações Hospedeiro-Patógeno , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas de Ligação a Fosfato/genética , Proteínas de Ligação a Fosfato/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA