Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 485
Filtrar
1.
Invest Ophthalmol Vis Sci ; 65(6): 13, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38848078

RESUMEN

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.


Asunto(s)
Candida albicans , Candidiasis , Modelos Animales de Enfermedad , Infecciones Fúngicas del Ojo , Proteínas de la Membrana , Nucleotidiltransferasas , Transducción de Señal , Animales , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/genética , Infecciones Fúngicas del Ojo/microbiología , Infecciones Fúngicas del Ojo/metabolismo , Ratones , Candida albicans/fisiología , Candidiasis/microbiología , Candidiasis/metabolismo , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Queratitis/microbiología , Queratitis/metabolismo , Western Blotting , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Femenino , Úlcera de la Córnea/microbiología , Úlcera de la Córnea/metabolismo , Inflamasomas/metabolismo
2.
Cell Mol Life Sci ; 81(1): 280, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38918243

RESUMEN

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.


Asunto(s)
Apoptosis , Candida albicans , Candidiasis , Proteínas de Unión al ADN , Macrófagos , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Animales , Macrófagos/metabolismo , Macrófagos/inmunología , Macrófagos/microbiología , Candidiasis/inmunología , Candidiasis/microbiología , Candidiasis/metabolismo , Candidiasis/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratones , Humanos , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Inflamasomas/metabolismo , Inmunidad Innata , Riñón/patología , Riñón/metabolismo , Riñón/microbiología
3.
Sci Rep ; 14(1): 14723, 2024 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-38926392

RESUMEN

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.


Asunto(s)
Biomarcadores , Antígenos CD36 , Candida albicans , Candidiasis , Exosomas , Macrófagos , Exosomas/metabolismo , Biomarcadores/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Macrófagos/inmunología , Antígenos CD36/metabolismo , Candidiasis/diagnóstico , Candidiasis/microbiología , Candidiasis/metabolismo , Candidiasis/inmunología , Humanos , Animales , Ratones
4.
PLoS Pathog ; 20(5): e1012225, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38739655

RESUMEN

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.


Asunto(s)
Biopelículas , Candida albicans , Candidiasis , Proteínas Fúngicas , Biopelículas/crecimiento & desarrollo , Candida albicans/metabolismo , Candida albicans/genética , Candida albicans/fisiología , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/genética , Animales , Candidiasis/microbiología , Candidiasis/metabolismo , Hifa/metabolismo , Ratones , Regulación Fúngica de la Expresión Génica , Ergosterol/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Mutación
5.
Open Biol ; 14(5): 230315, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38806144

RESUMEN

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.


Asunto(s)
Candida glabrata , Células Dendríticas , Antígeno de Macrófago-1 , Linfocitos T Reguladores , beta-Glucanos , Candida glabrata/metabolismo , Candida glabrata/patogenicidad , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , beta-Glucanos/metabolismo , beta-Glucanos/farmacología , Animales , Antígeno de Macrófago-1/metabolismo , Ratones , Lectinas Tipo C/metabolismo , Candidiasis/inmunología , Candidiasis/microbiología , Candidiasis/metabolismo , Ratones Endogámicos C57BL
6.
Cell Host Microbe ; 32(6): 964-979.e7, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38754418

RESUMEN

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.


Asunto(s)
Aterosclerosis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Candida albicans , Ceramidas , Transducción de Señal , Animales , Candida albicans/metabolismo , Aterosclerosis/microbiología , Aterosclerosis/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Ratones , Humanos , Ceramidas/metabolismo , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Masculino , Microbioma Gastrointestinal/fisiología , Intestinos/microbiología , Intestinos/patología , Disbiosis/microbiología , Femenino , Candidiasis/microbiología , Candidiasis/metabolismo
7.
Microbes Infect ; 26(4): 105305, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38296157

RESUMEN

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.


Asunto(s)
Candida albicans , Células Endoteliales , Glucólisis , Hígado , Candida albicans/inmunología , Células Endoteliales/metabolismo , Células Endoteliales/microbiología , Animales , Hígado/metabolismo , Hígado/microbiología , Quinasa Syk/metabolismo , Interleucina-6/metabolismo , Interleucina-6/genética , Ratones , Factor 88 de Diferenciación Mieloide/metabolismo , Inflamación/metabolismo , Perfilación de la Expresión Génica , Candidiasis/inmunología , Candidiasis/microbiología , Candidiasis/metabolismo
8.
Nat Microbiol ; 9(1): 95-107, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38168615

RESUMEN

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.


Asunto(s)
Candidiasis , Interferón Tipo I , Animales , Ratones , Candida albicans/patogenicidad , Proteínas Adaptadoras de Señalización CARD/metabolismo , Inmunidad Innata , Interferón Tipo I/metabolismo , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Transducción de Señal , Candidiasis/metabolismo , Candidiasis/patología
9.
PLoS Pathog ; 20(1): e1011902, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38166150

RESUMEN

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.


Asunto(s)
Candidiasis , Neutrófilos , Animales , Ratones , Antifúngicos , Candida albicans , Candidiasis/metabolismo , Candidiasis/microbiología , Citocinas/metabolismo , Infiltración Neutrófila , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas
10.
Int J Mol Sci ; 24(14)2023 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-37511508

RESUMEN

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.


Asunto(s)
Candida albicans , Candidiasis , Humanos , Candida albicans/genética , Redes Reguladoras de Genes , Células Endoteliales/metabolismo , Consenso , Candidiasis/metabolismo , Células Epiteliales/metabolismo , Proteínas Nucleares/genética
11.
Int J Clin Pract ; 2022: 5279323, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35832797

RESUMEN

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.


Asunto(s)
Candidiasis , Histatinas , Adolescente , Adulto , Antifúngicos/uso terapéutico , Candida , Candidiasis/tratamiento farmacológico , Candidiasis/metabolismo , Femenino , Histatinas/metabolismo , Humanos , Persona de Mediana Edad , Saliva/metabolismo , Adulto Joven
12.
Zhonghua Yi Xue Za Zhi ; 102(25): 1924-1930, 2022 Jul 05.
Artículo en Chino | MEDLINE | ID: mdl-35768392

RESUMEN

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.


Asunto(s)
Candida albicans , Candidiasis , Antifúngicos , Candida albicans/fisiología , Candidiasis/metabolismo , Células Epiteliales/metabolismo , Fluconazol/metabolismo , Fluconazol/farmacología , Factor Estimulante de Colonias de Granulocitos/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos
13.
J Microbiol ; 60(4): 402-410, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35157222

RESUMEN

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.


Asunto(s)
Lesión Pulmonar Aguda , Candida albicans , Candidiasis , MicroARNs , Proteína 1 Supresora de la Señalización de Citocinas , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/metabolismo , Animales , Candida albicans/genética , Candida albicans/metabolismo , Candidiasis/genética , Candidiasis/metabolismo , Candidiasis/microbiología , Regulación hacia Abajo , Inflamación/metabolismo , Inflamación/microbiología , MicroARNs/genética , MicroARNs/metabolismo , Proteína 1 Supresora de la Señalización de Citocinas/genética , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo
14.
Carbohydr Polym ; 283: 119178, 2022 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-35153023

RESUMEN

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.


Asunto(s)
Antifúngicos/farmacología , Candidiasis/tratamiento farmacológico , Quitosano/química , Fluconazol/farmacología , Liposomas/química , Nanopartículas/química , Adhesividad , Administración Bucal , Animales , Candida albicans/efectos de los fármacos , Candidiasis/metabolismo , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos/métodos , Glicéridos/química , Mucosa Bucal/metabolismo , Tamaño de la Partícula , Espectroscopía Infrarroja por Transformada de Fourier/métodos , Porcinos
15.
Bioengineered ; 13(2): 2513-2524, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35034584

RESUMEN

Antimicrobial peptides (AMPs) have proven to inhibit a variety of pathogens. Chromogranin A-N12 (CGA-N12) is a kind of AMP, and it is characterized by stable structure, high anti-Candida activity, and good safety. However, it remains unclear whether CGA-N12 could effectively inhibit the growth of Candida albicans (C. albicans). Colony forming assays were used to measure minimal inhibitory concentration (MIC), minimal fungicidal concentration (MFC), and time-kill curve. Disseminated C. albicans rabbit model was established to investigate the influence of CGA-N12 on histological damage. The protein and mRNA levels of suppressor of cytokine signaling 1 (SOCS1) after treatment were investigated. The MIC and MFC of CGA-N12 against C. albicans was 6 mg/mL. CGA-N12 considerably inhibited germ tube formation of C. albicans. The fungal load in the tissues and inflammatory factors in the serum were suppressed by CGA-N12. CGA-N12 significantly reduced the histological changes caused by C. albicans, and the protein and mRNA levels of SOCS1 were markedly inhibited. The inhibition effect of CGA-N12 on C. albicans and significant improvement of histological damage by CGA-N12 through microRNA-155/SOCS1 axis were proved in this study. This study proposes a novel therapeutic strategy for the treatment and prevention of C. albicans.Abbreviations: AMPs: Antimicrobial peptides; MIC: Minimal inhibitory concentration; MFC: Minimal fungicidal concentration; AIDS: Acquired immune deficiency syndrome; PBS: Phosphate buffer saline; FBS: Fetal bovine serum; ROS: Reactive oxygen species; CFU: Colony formation unit; CGA: Chromogranin A; SOCS1: Suppressor of cytokine signaling 1; SDA: Sabouraud Dextrose Agar; GRAVY: Grand average of hydropathicity; C. parapsilosis: Candida parapsilosis; C. albicans: Candida albicans.


Asunto(s)
Péptidos Antimicrobianos/farmacología , Candida albicans/metabolismo , Candidiasis/metabolismo , Cromogranina A/farmacología , MicroARNs/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Animales , Péptidos Antimicrobianos/química , Candidiasis/tratamiento farmacológico , Cromogranina A/química , Conejos
16.
Gut Microbes ; 14(1): 2004798, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35086419

RESUMEN

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.


Asunto(s)
Autofagia , Candida albicans/fisiología , Candidiasis/microbiología , Membrana Celular/microbiología , Células Epiteliales/microbiología , Proteína 5 Relacionada con la Autofagia/genética , Proteína 5 Relacionada con la Autofagia/metabolismo , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Candida albicans/genética , Candidiasis/genética , Candidiasis/metabolismo , Candidiasis/fisiopatología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Microbioma Gastrointestinal , Interacciones Huésped-Patógeno , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de Unión a Fosfato/genética , Proteínas de Unión a Fosfato/metabolismo
17.
J Biol Chem ; 298(2): 101593, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35051415

RESUMEN

Invasive candidiasis poses a major healthcare threat. The human opportunistic fungal pathogen Candida glabrata, which causes mucosal and deep-seated infections, is armed with distinct virulence attributes, including a family of 11 glycosylphosphatidylinositol-linked aspartyl proteases, CgYapsins. Here, we have profiled total membrane proteomes of the C. glabrata wildtype and 11 proteases-deficient strain, Cgyps1-11Δ, by mass spectrometry analysis and uncovered a novel role for fungal yapsins in glucose sensing and homeostasis. Furthermore, through label-free quantitative membrane proteome analysis, we showed differential abundance of 42% of identified membrane proteins, with electron transport chain and glycolysis proteins displaying lower and higher abundance in Cgyps1-11Δ cells, compared with wildtype cells, respectively. We also demonstrated elevated glucose uptake and upregulation of genes that code for the low-glucose sensor CgSnf3, transcriptional regulators CgMig1 and CgRgt1, and hexose transporter CgHxt2/10 in the Cgyps1-11Δ mutant. We further elucidated a potential underlying mechanism through genetic and transcript measurement analysis under low- and high-glucose conditions and found CgSNF3 deletion to rescue high glucose uptake and attenuated growth of the Cgyps1-11Δ mutant in YPD medium, thereby linking CgYapsins with regulation of the CgSnf3-dependent low-glucose sensing pathway. Last, high ethanol production, diminished mitochondrial membrane potential, and elevated susceptibility to oxidative phosphorylation inhibitors point toward increased fermentative and decreased respiratory metabolism in the Cgyps1-11Δ mutant. Altogether, our findings revealed new possible glucose metabolism-regulatory roles for putative cell surface-associated CgYapsins and advanced our understanding of fungal carbohydrate homeostasis mechanisms.


Asunto(s)
Proteasas de Ácido Aspártico , Candidiasis , Ácido Aspártico Endopeptidasas/metabolismo , Proteasas de Ácido Aspártico/genética , Proteasas de Ácido Aspártico/metabolismo , Candida glabrata , Candidiasis/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Glucosa/metabolismo , Homeostasis , Humanos
18.
PLoS Pathog ; 18(1): e1010192, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34995333

RESUMEN

Candida albicans is a major opportunistic pathogen of humans. It can grow as morphologically distinct yeast, pseudohyphae and hyphae, and the ability to switch reversibly among different forms is critical for its virulence. The relationship between morphogenesis and innate immune recognition is not quite clear. Dectin-1 is a major C-type lectin receptor that recognizes ß-glucan in the fungal cell wall. C. albicans ß-glucan is usually masked by the outer mannan layer of the cell wall. Whether and how ß-glucan masking is differentially regulated during hyphal morphogenesis is not fully understood. Here we show that the endo-1,3-glucanase Eng1 is differentially expressed in yeast, and together with Yeast Wall Protein 1 (Ywp1), regulates ß-glucan exposure and Dectin-1-dependent immune activation of macrophage by yeast cells. ENG1 deletion results in enhanced Dectin-1 binding at the septa of yeast cells; while eng1 ywp1 yeast cells show strong overall Dectin-1 binding similar to hyphae of wild-type and eng1 mutants. Correlatively, hyphae of wild-type and eng1 induced similar levels of cytokines in macrophage. ENG1 expression and Eng1-mediated ß-glucan trimming are also regulated by antifungal drugs, lactate and N-acetylglucosamine. Deletion of ENG1 modulates virulence in the mouse model of hematogenously disseminated candidiasis in a Dectin-1-dependent manner. The eng1 mutant exhibited attenuated lethality in male mice, but enhanced lethality in female mice, which was associated with a stronger renal immune response and lower fungal burden. Thus, Eng1-regulated ß-glucan exposure in yeast cells modulates the balance between immune protection and immunopathogenesis during disseminated candidiasis.


Asunto(s)
Candida albicans/patogenicidad , Candidiasis/inmunología , Glucano Endo-1,3-beta-D-Glucosidasa/metabolismo , Virulencia/fisiología , beta-Glucanos/inmunología , Animales , Candida albicans/inmunología , Candida albicans/metabolismo , Candidiasis/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , beta-Glucanos/metabolismo
19.
Bioengineered ; 13(1): 253-267, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34709974

RESUMEN

Microorganisms mainly exist in the form of biofilm in nature. Biofilm can contaminate food and drinking water system, as well as cause chronic wound infections, thereby posing a potential threat to public health safety. In the last two decades, researchers have made efforts to investigate the genetic contributors control different stages of biofilm development (adherence, initiation, maturation, and dispersal). As an opportunistic pathogen, C. albicans causes severe superficial or systemic infections with high morbidity and mortality under conditions of immune dysfunction. It has been reported that 80% of C. albicans infections are directly or indirectly associated with biofilm formation on host or abiotic surfaces including indwelling medical devices, resulting in high morbidity and mortality. Significantly, the outcome of C. albicans biofilm development includes enhanced invasion, exacerbated inflammatory responses and intrinsic resistance to antimicrobial chemotherapy. Thus, this review aimed at providing a comprehensive overview of the regulatory network controls microbial biofilm development, with C. albicans as a representative, served as reference for therapeutic targets.


Asunto(s)
Antifúngicos/uso terapéutico , Biopelículas , Candida albicans/fisiología , Candidiasis , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Candidiasis/tratamiento farmacológico , Candidiasis/metabolismo , Candidiasis/mortalidad , Proteínas Fúngicas/metabolismo , Humanos
20.
Front Immunol ; 12: 797550, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34956233

RESUMEN

Successful pathogens require metabolic flexibility to adapt to diverse host niches. The presence of co-infecting or commensal microorganisms at a given infection site can further influence the metabolic processes required for a pathogen to cause disease. The Gram-positive bacterium Staphylococcus aureus and the polymorphic fungus Candida albicans are microorganisms that asymptomatically colonize healthy individuals but can also cause superficial infections or severe invasive disease. Due to many shared host niches, S. aureus and C. albicans are frequently co-isolated from mixed fungal-bacterial infections. S. aureus and C. albicans co-infection alters microbial metabolism relative to infection with either organism alone. Metabolic changes during co-infection regulate virulence, such as enhancing toxin production in S. aureus or contributing to morphogenesis and cell wall remodeling in C. albicans. C. albicans and S. aureus also form polymicrobial biofilms, which have greater biomass and reduced susceptibility to antimicrobials relative to mono-microbial biofilms. The S. aureus and C. albicans metabolic programs induced during co-infection impact interactions with host immune cells, resulting in greater microbial survival and immune evasion. Conversely, innate immune cell sensing of S. aureus and C. albicans triggers metabolic changes in the host cells that result in an altered immune response to secondary infections. In this review article, we discuss the metabolic programs that govern host-pathogen interactions during S. aureus and C. albicans co-infection. Understanding C. albicans-S. aureus interactions may highlight more general principles of how polymicrobial interactions, particularly fungal-bacterial interactions, shape the outcome of infectious disease. We focus on how co-infection alters microbial metabolism to enhance virulence and how infection-induced changes to host cell metabolism can impact a secondary infection.


Asunto(s)
Candida albicans/fisiología , Candidiasis/metabolismo , Coinfección/metabolismo , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/fisiología , Adaptación Fisiológica , Animales , Biopelículas , Humanos , Interacciones Microbianas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA