Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Front Cell Infect Microbiol ; 13: 1254919, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37808914

RESUMO

Mucormycosis (MCR) is an emerging and frequently lethal fungal infection caused by the Mucorales family, with Rhizopus, Mucor, and Lichtheimia, accounting for > 90% of all cases. MCR is seen in patients with severe immunosuppression such as those with hematologic malignancy or transplantation, Diabetes Mellitus (DM) and diabetic ketoacidosis (DKA) and immunocompetent patients with severe wounds. The recent SARS COV2 epidemy in India has resulted in a tremendous increase in MCR cases, typically seen in the setting of uncontrolled DM and corticosteroid use. In addition to the diversity of affected hosts, MCR has pleiotropic clinical presentations, with rhino-orbital/rhino-cerebral, sino-pulmonary and necrotizing cutaneous forms being the predominant manifestations. Major insights in MCR pathogenesis have brought into focus the host receptors (GRP78) and signaling pathways (EGFR activation cascade) as well as the adhesins used by Mucorales for invasion. Furthermore, studies have expanded on the importance of iron availability and the complex regulation of iron homeostasis, as well as the pivotal role of mycotoxins as key factors for tissue invasion. The molecular toolbox to study Mucorales pathogenesis remains underdeveloped, but promise is brought by RNAi and CRISPR/Cas9 approaches. Important recent advancements have been made in early, culture-independent molecular diagnosis of MCR. However, development of new potent antifungals against Mucorales remains an unmet need. Therapy of MCR is multidisciplinary and requires a high index of suspicion for initiation of early Mucorales-active antifungals. Reversal of underlying immunosuppression, if feasible, rapid DKA correction and in selected patients, surgical debulking are crucial for improved outcomes.


Assuntos
COVID-19 , Diabetes Mellitus , Mucorales , Mucormicose , Humanos , Mucormicose/diagnóstico , Mucormicose/tratamento farmacológico , Antifúngicos/uso terapêutico , Ferro
2.
PLoS Pathog ; 19(8): e1011579, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37611070

RESUMO

Fungal invasion of the oral epithelium is central to the pathogenesis of oropharyngeal candidiasis (OPC). Candida albicans invades the oral epithelium by receptor-induced endocytosis but this process is incompletely understood. We found that C. albicans infection of oral epithelial cells induces c-Met to form a multi-protein complex with E-cadherin and the epidermal growth factor receptor (EGFR). E-cadherin is necessary for C. albicans to activate both c-Met and EGFR and to induce the endocytosis of C. albicans. Proteomics analysis revealed that c-Met interacts with C. albicans Hyr1, Als3 and Ssa1. Both Hyr1 and Als3 are required for C. albicans to stimulate c-Met and EGFR in oral epithelial cells in vitro and for full virulence during OPC in mice. Treating mice with small molecule inhibitors of c-Met and EGFR ameliorates OPC, demonstrating the potential therapeutic efficacy of blocking these host receptors for C. albicans.


Assuntos
Candida albicans , Candidíase Bucal , Animais , Camundongos , Membrana Celular , Receptores ErbB , Caderinas , Células Epiteliais
3.
Microbiol Spectr ; 11(4): e0008423, 2023 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-37255456

RESUMO

Invasive aspergillosis is initiated when Aspergillus fumigatus adheres to and invades the pulmonary epithelial cells that line the airways and alveoli. To gain deeper insight into how pulmonary epithelial cells respond to A. fumigatus invasion, we used transcriptome sequencing (RNA-seq) to determine the transcriptional response of the A549 type II alveolar epithelial cell line to infection with strains CEA10 and Af293, two clinical isolates of A. fumigatus. Upstream regulator analysis of the data indicated that while both strains activated virtually identical host cell signaling pathways after 16 h of infection, only strain CEA10 activated these pathways after 6 h of infection. Many of the pathways that were predicted to be activated by A. fumigatus, including the tumor necrosis factor (TNF), interleukin-1α (IL-1α), IL-1ß, IL-17A, Toll-like receptor 2 (TLR2), and TLR4 pathways, are known to be critical for the host defense against this fungus. We also found that the platelet-derived growth factor BB (PDGF BB) and progesterone receptor (PGR) pathways were activated by A. fumigatus. Using pharmacologic inhibitors, we determined that blocking the PDGF receptor or PGR inhibited the endocytosis of both strains of A. fumigatus in an additive manner. Both the PDGF BB and PGR pathways are also predicted to be activated by infection of A549 cells with other molds, such as Rhizopus delemar and Rhizopus oryzae. Thus, these pathways may represent a common response of pulmonary epithelial cells to mold infection. IMPORTANCE Invasive aspergillosis is a deadly invasive fungal infection that initiates when Aspergillus fumigatus spores are inhaled and come into contact with the epithelial cells that line the airways and alveoli. Understanding this fungus-host interaction is important for the development of novel therapeutics. To gain a deeper understanding of how these airway epithelial cells respond to A. fumigatus during infection, we used RNA-seq to determine the transcriptional response of alveolar epithelial cells to infection with two different clinical isolates of A. fumigatus. Our analysis identified new host response pathways that have not previously been tied to infection with A. fumigatus. Pharmacological inhibition of two of these pathways inhibited the ability of A. fumigatus to invade airway epithelial cells. These two pathways are also predicted to be activated by infection with other filamentous fungi. Thus, these pathways may represent a common response of alveolar epithelial cells to mold infection.


Assuntos
Aspergilose , Aspergillus fumigatus , Humanos , Becaplermina , Aspergilose/microbiologia , Células Epiteliais/microbiologia , Pulmão/microbiologia
4.
Nat Microbiol ; 6(3): 313-326, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33462434

RESUMO

Fungi of the order Mucorales cause mucormycosis, a lethal infection with an incompletely understood pathogenesis. We demonstrate that Mucorales fungi produce a toxin, which plays a central role in virulence. Polyclonal antibodies against this toxin inhibit its ability to damage human cells in vitro and prevent hypovolemic shock, organ necrosis and death in mice with mucormycosis. Inhibition of the toxin in Rhizopus delemar through RNA interference compromises the ability of the fungus to damage host cells and attenuates virulence in mice. This 17 kDa toxin has structural and functional features of the plant toxin ricin, including the ability to inhibit protein synthesis through its N-glycosylase activity, the existence of a motif that mediates vascular leak and a lectin sequence. Antibodies against the toxin inhibit R. delemar- or toxin-mediated vascular permeability in vitro and cross react with ricin. A monoclonal anti-ricin B chain antibody binds to the toxin and also inhibits its ability to cause vascular permeability. Therefore, we propose the name 'mucoricin' for this toxin. Not only is mucoricin important in the pathogenesis of mucormycosis but our data suggest that a ricin-like toxin is produced by organisms beyond the plant and bacterial kingdoms. Importantly, mucoricin should be a promising therapeutic target.


Assuntos
Mucorales/patogenicidade , Mucormicose/patologia , Micotoxinas/metabolismo , Ricina/metabolismo , Animais , Antitoxinas/imunologia , Antitoxinas/farmacologia , Antitoxinas/uso terapêutico , Apoptose , Permeabilidade Capilar , Células Cultivadas , Reações Cruzadas , Humanos , Hifas/química , Hifas/patogenicidade , Lectinas/metabolismo , Camundongos , Mucorales/química , Mucorales/classificação , Mucorales/genética , Mucormicose/microbiologia , Mucormicose/prevenção & controle , Micotoxinas/química , Micotoxinas/genética , Micotoxinas/imunologia , Necrose , Interferência de RNA , Rhizopus/química , Rhizopus/genética , Rhizopus/patogenicidade , Proteínas Inativadoras de Ribossomos/metabolismo , Ricina/química , Ricina/imunologia , Virulência/efeitos dos fármacos , Virulência/genética
5.
Nat Commun ; 11(1): 3387, 2020 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-32636417

RESUMO

Biosynthesis of glycosylphosphatidylinositol (GPI) is required for anchoring proteins to the plasma membrane, and is essential for the integrity of the fungal cell wall. Here, we use a reporter gene-based screen in Saccharomyces cerevisiae for the discovery of antifungal inhibitors of GPI-anchoring of proteins, and identify the oligocyclopropyl-containing natural product jawsamycin (FR-900848) as a potent hit. The compound targets the catalytic subunit Spt14 (also referred to as Gpi3) of the fungal UDP-glycosyltransferase, the first step in GPI biosynthesis, with good selectivity over the human functional homolog PIG-A. Jawsamycin displays antifungal activity in vitro against several pathogenic fungi including Mucorales, and in vivo in a mouse model of invasive pulmonary mucormycosis due to Rhyzopus delemar infection. Our results provide a starting point for the development of Spt14 inhibitors for treatment of invasive fungal infections.


Assuntos
Antifúngicos/farmacologia , Glicosiltransferases/antagonistas & inibidores , Policetídeos/farmacologia , Proteínas de Saccharomyces cerevisiae/antagonistas & inibidores , Animais , Proliferação de Células , Modelos Animais de Doenças , Fermentação , Genes Reporter , Glicosilfosfatidilinositóis/biossíntese , Células HCT116 , Células Hep G2 , Humanos , Concentração de Íons de Hidrogênio , Concentração Inibidora 50 , Células K562 , Pulmão/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Mucorales , Família Multigênica , Rhizopus , Saccharomyces cerevisiae
6.
mBio ; 11(3)2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32487760

RESUMO

Mucormycosis, caused by Rhizopus species, is a life-threatening fungal infection that occurs in patients immunocompromised by diabetic ketoacidosis (DKA), cytotoxic chemotherapy, immunosuppressive therapy, hematologic malignancies, or severe trauma. Inhaled Rhizopus spores cause pulmonary infections in patients with hematologic malignancies, while patients with DKA are much more prone to rhinoorbital/cerebral mucormycosis. Here, we show that Rhizopus delemar interacts with glucose-regulated protein 78 (GRP78) on nasal epithelial cells via its spore coat protein CotH3 to invade and damage the nasal epithelial cells. Expression of the two proteins is significantly enhanced by high glucose, iron, and ketone body levels (hallmark features of DKA), potentially leading to frequently lethal rhinoorbital/cerebral mucormycosis. In contrast, R. delemar CotH7 recognizes integrin ß1 as a receptor on alveolar epithelial cells, causing the activation of epidermal growth factor receptor (EGFR) and leading to host cell invasion. Anti-integrin ß1 antibodies inhibit R. delemar invasion of alveolar epithelial cells and protect mice from pulmonary mucormycosis. Our results show that R. delemar interacts with different mammalian receptors depending on the host cell type. Susceptibility of patients with DKA primarily to rhinoorbital/cerebral disease can be explained by host factors typically present in DKA and known to upregulate CotH3 and nasal GRP78, thereby trapping the fungal cells within the rhinoorbital milieu, leading to subsequent invasion and damage. Our studies highlight that mucormycosis pathogenesis can potentially be overcome by the development of novel customized therapies targeting niche-specific host receptors or their respective fungal ligands.IMPORTANCE Mucormycosis caused by Rhizopus species is a fungal infection with often fatal prognosis. Inhalation of spores is the major route of entry, with nasal and alveolar epithelial cells among the first cells that encounter the fungi. In patients with hematologic malignancies or those undergoing cytotoxic chemotherapy, Rhizopus causes pulmonary infections. On the other hand, DKA patients predominantly suffer from rhinoorbital/cerebral mucormycosis. The reason for such disparity in disease types by the same fungus is not known. Here, we show that the unique susceptibility of DKA subjects to rhinoorbital/cerebral mucormycosis is likely due to specific interaction between nasal epithelial cell GRP78 and fungal CotH3, the expression of which increases in the presence of host factors present in DKA. In contrast, pulmonary mucormycosis is initiated via interaction of inhaled spores expressing CotH7 with integrin ß1 receptor, which activates EGFR to induce fungal invasion of host cells. These results introduce a plausible explanation for disparate disease manifestations in DKA versus those in hematologic malignancy patients and provide a foundation for development of therapeutic interventions against these lethal forms of mucormycosis.


Assuntos
Células Epiteliais/microbiologia , Proteínas de Choque Térmico/genética , Interações Hospedeiro-Patógeno , Infecções Fúngicas Invasivas/microbiologia , Mucormicose/microbiologia , Receptores de Vitronectina/genética , Rhizopus/patogenicidade , Células A549 , Células Epiteliais Alveolares/microbiologia , Células Epiteliais Alveolares/patologia , Animais , Linhagem Celular , Cetoacidose Diabética/complicações , Cetoacidose Diabética/microbiologia , Chaperona BiP do Retículo Endoplasmático , Células Epiteliais/patologia , Receptores ErbB/genética , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Nariz/citologia , Virulência
7.
Nat Commun ; 9(1): 5015, 2018 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-30467313

RESUMO

The original version of this Article contained an error in the spelling of the author Emilien Etienne, which was incorrectly given as Emilien Ettiene. These errors have now been corrected in both the PDF and HTML versions of the Article.

8.
mBio ; 9(4)2018 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-30108171

RESUMO

Mucormycosis is a life-threatening, invasive fungal infection that is caused by various species belonging to the order Mucorales. Rhizopus species are the most common cause of the disease, responsible for approximately 70% of all cases of mucormycosis. During pulmonary mucormycosis, inhaled Rhizopus spores must adhere to and invade airway epithelial cells in order to establish infection. The molecular mechanisms that govern this interaction are poorly understood. We performed an unbiased survey of the host transcriptional response during early stages of Rhizopus arrhizus var. delemar (R. delemar) infection in a murine model of pulmonary mucormycosis using transcriptome sequencing (RNA-seq). Network analysis revealed activation of the host's epidermal growth factor receptor (EGFR) signaling. Consistent with the RNA-seq results, EGFR became phosphorylated upon in vitro infection of human alveolar epithelial cells with several members of the Mucorales, and this phosphorylated, activated form of EGFR colocalized with R. delemar spores. Inhibition of EGFR signaling with cetuximab or gefitinib, specific FDA-approved inhibitors of EGFR, significantly reduced the ability of R. delemar to invade and damage airway epithelial cells. Furthermore, gefitinib treatment significantly prolonged survival of mice with pulmonary mucormycosis, reduced tissue fungal burden, and attenuated the activation of EGFR in response to pulmonary mucormycosis. These results indicate EGFR represents a novel host target to block invasion of alveolar epithelial cells by R. delemar, and inhibition of EGFR signaling provides a novel approach for treating mucormycosis by repurposing an FDA-approved drug.IMPORTANCE Mucormycosis is an increasingly common, highly lethal fungal infection with very limited treatment options. Using a combination of in vivo animal models, transcriptomics, cell biology, and pharmacological approaches, we have demonstrated that Mucorales fungi activate EGFR signaling to induce fungal uptake into airway epithelial cells. Inhibition of EGFR signaling with existing FDA-approved drugs significantly increased survival following R. arrhizus var. delemar infection in mice. This study enhances our understanding of how Mucorales fungi invade host cells during the establishment of pulmonary mucormycosis and provides a proof-of-concept for the repurposing of FDA-approved drugs that target EGFR function.


Assuntos
Receptores ErbB/antagonistas & inibidores , Interações Hospedeiro-Patógeno , Pulmão/microbiologia , Mucormicose/prevenção & controle , Células A549 , Animais , Cetuximab/farmacologia , Modelos Animais de Doenças , Receptores ErbB/metabolismo , Gefitinibe/farmacologia , Redes Reguladoras de Genes , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Mucormicose/microbiologia , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Rhizopus/efeitos dos fármacos , Rhizopus/patogenicidade , Análise de Sequência de RNA , Transdução de Sinais/efeitos dos fármacos
9.
Nat Commun ; 9(1): 3333, 2018 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-30127354

RESUMO

Mucormycosis is a life-threatening respiratory fungal infection predominantly caused by Rhizopus species. Mucormycosis has incompletely understood pathogenesis, particularly how abnormalities in iron metabolism compromise immune responses. Here we show how, as opposed to other filamentous fungi, Rhizopus spp. establish intracellular persistence inside alveolar macrophages (AMs). Mechanistically, lack of intracellular swelling of Rhizopus conidia results in surface retention of melanin, which induces phagosome maturation arrest through inhibition of LC3-associated phagocytosis. Intracellular inhibition of Rhizopus is an important effector mechanism, as infection of immunocompetent mice with swollen conidia, which evade phagocytosis, results in acute lethality. Concordantly, AM depletion markedly increases susceptibility to mucormycosis. Host and pathogen transcriptomics, iron supplementation studies, and genetic manipulation of iron assimilation of fungal pathways demonstrate that iron restriction inside macrophages regulates immunity against Rhizopus. Our findings shed light on the pathogenetic mechanisms of mucormycosis and reveal the role of macrophage-mediated nutritional immunity against filamentous fungi.


Assuntos
Interações Hospedeiro-Patógeno , Ferro/metabolismo , Pulmão/microbiologia , Macrófagos Alveolares/metabolismo , Rhizopus/fisiologia , Animais , Parede Celular/metabolismo , Regulação da Expressão Gênica , Macrófagos Alveolares/ultraestrutura , Melaninas/metabolismo , Camundongos Endogâmicos C57BL , Viabilidade Microbiana , Modelos Biológicos , Mucormicose/genética , Mucormicose/microbiologia , Mucormicose/patologia , Fagossomos/metabolismo , Fagossomos/ultraestrutura , Rhizopus/crescimento & desenvolvimento , Esporos Fúngicos/fisiologia
10.
Curr Opin Microbiol ; 40: 40-45, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29107938

RESUMO

Mucormycosis is a fungal infection with fulminant angioinvasion leading to high morbidity and mortality in susceptible individuals. The major predisposing conditions are uncontrolled diabetes, neutropenia, malignancies, receipt of a transplant and traumatic injury [1]. Over the past decade, mucormycosis has become an emerging fungal infection due to the increase in patient groups presenting with these pre-disposing conditions and our medical advances in diagnosing the infection [2-4]. Yet, we currently lack clinical interventions to treat mucormycosis effectively. This in turn is due to a lack of understanding of mucormycosis pathogenesis. Here, we discuss our current understanding of selected aspects of interactions at the mucormycete-host interface. We will highlight open questions that might guide future research directions for investigations into the pathogenesis of mucormycosis and potential innovative therapeutic approaches.


Assuntos
Interações Hospedeiro-Patógeno , Mucorales/fisiologia , Mucormicose/microbiologia , Mucormicose/fisiopatologia , Animais , Humanos , Mucorales/genética
11.
Nat Microbiol ; 2: 16211, 2016 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-27841851

RESUMO

Aspergillus fumigatus is an opportunistic fungal pathogen that invades pulmonary epithelial cells and vascular endothelial cells by inducing its own endocytosis, but the mechanism by which this process occurs is poorly understood. Here, we show that the thaumatin-like protein CalA is expressed on the surface of the A. fumigatus cell wall, where it mediates invasion of epithelial and endothelial cells. CalA induces endocytosis in part by interacting with integrin α5ß1 on host cells. In corticosteroid-treated mice, a ΔcalA deletion mutant has significantly attenuated virulence relative to the wild-type strain, as manifested by prolonged survival, reduced pulmonary fungal burden and decreased pulmonary invasion. Pretreatment with an anti-CalA antibody improves survival of mice with invasive pulmonary aspergillosis, demonstrating the potential of CalA as an immunotherapeutic target. Thus, A. fumigatus CalA is an invasin that interacts with integrin α5ß1 on host cells, induces endocytosis and enhances virulence.


Assuntos
Aspergillus fumigatus/fisiologia , Aspergillus fumigatus/patogenicidade , Endocitose , Proteínas Fúngicas/metabolismo , Interações Hospedeiro-Patógeno , Integrina alfa5beta1/metabolismo , Células A549 , Animais , Contagem de Colônia Microbiana , Modelos Animais de Doenças , Células Epiteliais/microbiologia , Células Epiteliais/fisiologia , Proteínas Fúngicas/genética , Deleção de Genes , Humanos , Pulmão/microbiologia , Camundongos , Ligação Proteica , Aspergilose Pulmonar/microbiologia , Aspergilose Pulmonar/patologia , Análise de Sobrevida , Virulência , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
12.
J Infect Dis ; 214(1): 114-21, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-26984141

RESUMO

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.


Assuntos
Antifúngicos/farmacologia , Antifúngicos/uso terapêutico , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Mucormicose/tratamento farmacológico , Rhizopus/efeitos dos fármacos , Virulência/efeitos dos fármacos , Animais , Atorvastatina/farmacologia , Atorvastatina/uso terapêutico , Dípteros/efeitos dos fármacos , Chaperona BiP do Retículo Endoplasmático , Feminino , Humanos , Lovastatina/farmacologia , Lovastatina/uso terapêutico , Camundongos , Testes de Sensibilidade Microbiana , Sinvastatina/farmacologia , Sinvastatina/uso terapêutico , Esporos Fúngicos/efeitos dos fármacos , Texas
13.
PLoS Pathog ; 11(5): e1004842, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25974051

RESUMO

Dialysis patients with chronic renal failure receiving deferoxamine for treating iron overload are uniquely predisposed for mucormycosis, which is most often caused by Rhizopus oryzae. Although the deferoxamine siderophore is not secreted by Mucorales, previous studies established that Rhizopus species utilize iron from ferrioxamine (iron-rich form of deferoxamine). Here we determined that the CBS domain proteins of Fob1 and Fob2 act as receptors on the cell surface of R. oryzae during iron uptake from ferrioxamine. Fob1 and Fob2 cell surface expression was induced in the presence of ferrioxamine and bound radiolabeled ferrioxamine. A R. oryzae strain with targeted reduced Fob1/Fob2 expression was impaired for iron uptake, germinating, and growing on medium with ferrioxamine as the sole source of iron. This strain also exhibited reduced virulence in a deferoxamine-treated, but not the diabetic ketoacidotic (DKA), mouse model of mucormycosis. The mechanism by which R. oryzae obtains iron from ferrioxamine involves the reductase/permease uptake system since the growth on ferrioxamine supplemented medium is associated with elevated reductase activity and the use of the ferrous chelator bathophenanthroline disulfonate abrogates iron uptake and growth on medium supplemented with ferrioxamine as a sole source of iron. Finally, R. oryzae mutants with reduced copies of the high affinity iron permease (FTR1) or with decreased FTR1 expression had an impaired iron uptake from ferrioxamine in vitro and reduced virulence in the deferoxamine-treated mouse model of mucormycosis. These two receptors appear to be conserved in Mucorales, and can be the subject of future novel therapy to maintain the use of deferoxamine for treating iron-overload.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Desferroxamina/metabolismo , Compostos Férricos/metabolismo , Ferro/metabolismo , Glicoproteínas de Membrana/metabolismo , Mucormicose/tratamento farmacológico , Rhizopus/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Fatores de Virulência/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Quelantes de Ferro/farmacologia , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Mucormicose/microbiologia , Sideróforos/metabolismo
14.
Virulence ; 6(2): 121-6, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25830548

RESUMO

Mortality associated with mucormycosis remains high despite current antifungals. Iron-starvation strategies have been shown to have promising activity against Mucorales. We hypothesized that iron starvation enhances apoptosis in Rhizopus oryzae. Apoptosis was characterized in R. oryzae transformed with RNAi plasmid targeting FTR1 expression (iron permease mutant) or empty plasmid grown in iron rich (0.125% FeCl3) and iron depleted media (YNB+1mM ferrozine and 1 mM ascorbic acid). Increased apoptosis was observed with dihydrorhodamine-123 and rhodamine-123 staining in the iron starved mutant FTR1 when compared to empty plasmid, followed by increased extracellular ATP levels. In addition, DNA fragmentation and metacaspase activity were prominent in FTR1. In contrast, Rhizopus strains grown in iron-rich medium displayed minimal apoptosis. Our results demonstrate a metacaspase dependent apoptotic process in iron deprived condition and further support the role of iron starvation strategies as an adjunct treatment for mucormycosis, a mechanism by which iron starvation affects R. oryzae.


Assuntos
Apoptose , Ferro/metabolismo , Rhizopus/genética , Rhizopus/fisiologia , Caspases/metabolismo , Meios de Cultura , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Potencial da Membrana Mitocondrial , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Testes de Sensibilidade Microbiana , Mutação , Espécies Reativas de Oxigênio/metabolismo , Rhizopus/ultraestrutura
15.
Artigo em Inglês | MEDLINE | ID: mdl-25377138

RESUMO

In recent years, we have seen an increase in the number of immunocompromised cohorts as a result of infections and/or medical conditions, which has resulted in an increased incidence of fungal infections. Although rare, the incidence of infections caused by fungi belonging to basal fungal lineages is also continuously increasing. Basal fungal lineages diverged at an early point during the evolution of the fungal lineage, in which, in a simplified four-phylum fungal kingdom, Zygomycota and Chytridiomycota belong to the basal fungi, distinguishing them from Ascomycota and Basidiomycota. Currently there are no known human infections caused by fungi in Chytridiomycota; only Zygomycotan fungi are known to infect humans. Hence, infections caused by zygomycetes have been called zygomycosis, and the term "zygomycosis" is often used as a synonym for "mucormycosis." In the four-phylum fungal kingdom system, Zygomycota is classified mainly based on morphology, including the ability to form coenocytic (aseptated) hyphae and zygospores (sexual spores). In the Zygomycota, there are 10 known orders, two of which, the Mucorales and Entomophthorales, contain species that can infect humans, and the infection has historically been known as zygomycosis. However, recent multilocus sequence typing analyses (the fungal tree of life [AFTOL] project) revealed that the Zygomycota forms not a monophyletic clade but instead a polyphyletic clade, whereas Ascomycota and Basidiomycota are monophyletic. Thus, the term "zygomycosis" needed to be further specified, resulting in the terms "mucormycosis" and "entomophthoramycosis." This review covers these two different types of fungal infections.


Assuntos
Conidiobolus/patogenicidade , Entomophthorales/patogenicidade , Mucorales/patogenicidade , Mucormicose/microbiologia , Zigomicose/microbiologia , Anfotericina B/uso terapêutico , Animais , Humanos , Hospedeiro Imunocomprometido , Camundongos , Mucormicose/tratamento farmacológico , Iodeto de Potássio/uso terapêutico , Ensaios Clínicos Controlados Aleatórios como Assunto , Zigomicose/tratamento farmacológico
16.
Mycoses ; 57 Suppl 3: 13-7, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25178879

RESUMO

Mucormycosis is a fungal infection caused by organisms belonging to the order Mucorales. Although considered uncommon, mucormycosis has been steadily increasing in incidents for the last two decades. Mortality of the disease is unacceptably high despite antifungal therapy and surgical interventions. The lack of understanding of the pathogenesis of the disease and the absence of rapid diagnostic assay contribute to the poor prognosis of mucormycosis. The hyper susceptibility of patients with elevated available serum iron points to the critical role of the ability of Mucorales to acquire host iron as a critical virulence factor. Specifically patients with deferoxamine-therapy, hyperglycaemic with or without ketoacidosis, or other forms of acidosis are uniquely predisposed to mucormycosis. In this review, we discuss the molecular mechanisms of infection in these patient categories in an attempt to identify novel therapies for a disease with poor prognosis. Emphasis on the effect of glucose and free iron on host-pathogen interactions are also covered.


Assuntos
Interações Hospedeiro-Patógeno , Ferro/sangue , Mucormicose/tratamento farmacológico , Mucormicose/patologia , Antifúngicos/uso terapêutico , Chaperona BiP do Retículo Endoplasmático , Humanos , Hiperglicemia/complicações , Cetose/complicações , Mucorales , Fatores de Risco , Fatores de Virulência
17.
Infect Immun ; 81(7): 2528-35, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23630968

RESUMO

During hematogenously disseminated infection, blood-borne Candida albicans invades the endothelial cell lining of the vasculature to invade the deep tissues. Although the C. albicans Als3 invasin is critical for invasion and damage of endothelial cells in vitro, a C. albicans als3Δ/Δ mutant has normal virulence in the mouse model of disseminated infection. We hypothesized that the contribution of Als3 to virulence is obscured by the presence of additional C. albicans invasins. To elucidate the in vivo function of Als3, we heterologously expressed C. albicans ALS3 in Candida glabrata, a yeast that lacks a close ALS3 ortholog and has low virulence in mice. We found that following intravenous inoculation into mice, the ALS3-expressing strain preferentially trafficked to the brain, where it induced significantly elevated levels of myeloperoxidase, tumor necrosis factor, monocyte chemoattractant protein 1, and gamma interferon. Also, the ALS3-expressing strain had enhanced adherence to and invasion of human brain microvascular endothelial cells in vitro, demonstrating a potential mechanism for ALS3-mediated neurotropism. In addition, upon initiation of infection, the ALS3-expressing strain had increased trafficking to the cortex of the kidneys. With prolonged infection, this strain persisted in the kidneys at significantly higher levels than the control strain but did not induce an elevated inflammatory response. Finally, the ALS3-expressing strain had increased resistance to neutrophil killing in vitro. These results indicate that during disseminated infection, Als3 mediates initial trafficking to the brain and renal cortex and contributes to fungal persistence in the kidneys.


Assuntos
Candida albicans/patogenicidade , Candida glabrata/metabolismo , Proteínas Fúngicas/metabolismo , Regulação Fúngica da Expressão Gênica , Genes Fúngicos , Animais , Encéfalo/microbiologia , Encéfalo/patologia , Candida albicans/genética , Candida albicans/imunologia , Candida glabrata/genética , Candidíase/microbiologia , Adesão Celular , Linhagem Celular , Contagem de Colônia Microbiana , Endocitose , Proteínas Fúngicas/genética , Proteínas Fúngicas/imunologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/microbiologia , Interleucina-8/metabolismo , Córtex Renal/microbiologia , Córtex Renal/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/microbiologia , Peroxidase/metabolismo , Transporte Proteico
18.
Med Mycol ; 50(6): 611-8, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22435877

RESUMO

Optimal clinical care and clinical investigation of patients with mucormycosis are limited by absence of controlled trials, and absence of well-defined predictors of mortality or clinical response. The Deferasirox-AmBisome Therapy for mucormycosis (DEFEAT Mucor) study was the first randomized clinical trial conducted on patients with mucormycosis, and demonstrated that adjunctive deferasirox therapy did not improve outcomes of the disease. The current study describes clinical factors from the 20 patients enrolled to identify those associated with 90-day mortality of the 11 (55%) patients who died by day 90. Age, diabetes mellitus, transplant status, or antifungal therapy were not associated with mortality. However, active malignancy or neutropenia at enrollment were associated with increased mortality. Pulmonary infection was linked with lower Kaplan-Meier survival compared to non-pulmonary infection. Higher baseline serum concentrations of iron and ferritin were also associated with mortality. No patient who progressed clinically during the first 14 days of study therapy survived; however, many patients who clinically improved during that time did not survive to 90 days. In contrast, day 30 clinical response was predictive of 90-day survival. These factors may be useful in defining enrollment randomization stratification critieria for future clinical trials, and in supporting clinical care of patients with mucormycosis.


Assuntos
Pneumopatias/mortalidade , Mucor/patogenicidade , Mucormicose/mortalidade , Adulto , Idoso , Anfotericina B/uso terapêutico , Benzoatos/farmacologia , Deferasirox , Método Duplo-Cego , Ferritinas/sangue , Humanos , Ferro/sangue , Estimativa de Kaplan-Meier , Pneumopatias/tratamento farmacológico , Pneumopatias/microbiologia , Pessoa de Meia-Idade , Mucormicose/tratamento farmacológico , Mucormicose/microbiologia , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento , Triazóis/farmacologia
19.
Clin Infect Dis ; 54 Suppl 1: S16-22, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22247441

RESUMO

Mucormycosis is a life-threatening infection that occurs in patients who are immunocompromised because of diabetic ketoacidosis, neutropenia, organ transplantation, and/or increased serum levels of available iron. Because of the increasing prevalence of diabetes mellitus, cancer, and organ transplantation, the number of patients at risk for this deadly infection is increasing. Despite aggressive therapy, which includes disfiguring surgical debridement and frequently adjunctive toxic antifungal therapy, the overall mortality rate is high. New strategies to prevent and treat mucormycosis are urgently needed. Understanding the pathogenesis of mucormycosis and the host response to invading hyphae ultimately will provide targets for novel therapeutic interventions. In this supplement, we review the current knowledge about the virulence traits used by the most common etiologic agent of mucormycosis, Rhizopus oryzae. Because patients with elevated serum levels of available iron are uniquely susceptible to mucormycosis and these infections are highly angioinvasive, emphasis is placed on the ability of the organism to acquire iron from the host and on its interactions with endothelial cells lining blood vessels. Several promising therapeutic strategies in preclinical stages are identified.


Assuntos
Ferro/metabolismo , Mucormicose/patologia , Rhizopus/patogenicidade , Cetoacidose Diabética/metabolismo , Cetoacidose Diabética/microbiologia , Células Endoteliais/metabolismo , Células Endoteliais/microbiologia , Genes Fúngicos , Interações Hospedeiro-Patógeno , Humanos , Hospedeiro Imunocomprometido , Mucormicose/metabolismo , Mucormicose/microbiologia , Fagócitos/metabolismo , Fagócitos/patologia , Rhizopus/genética , Rhizopus/metabolismo , Fatores de Risco , Fatores de Virulência/metabolismo
20.
J Infect Dis ; 205(2): 337-47, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22124127

RESUMO

BACKGROUND: Activated transfected killer (ATAK) cells are immortal phagocytes transfected with a luminescence reporter that effectively treat lethal infections in neutropenic mice. Their in vivo trafficking, lifespan, and immunogenicity are unknown. METHODS: Mice were made neutropenic; infected or not with Staphylococcus aureus, Acinetobacter baumannii, Candida albicans, or Aspergillus fumigatus; and treated intraperitoneally with ATAK cells. Cell trafficking and lifespan were assessed by in vivo imaging and reverse transcription-polymerase chain reaction. RESULTS: In uninfected neutropenic mice, ATAK cells spread from the mesentery into visceral organs on days 1-3. Splenic accumulation of ATAK cells increased at day 1 after infection with S. aureus and A. baumannii, and kidney accumulation increased in mice infected with C. albicans. Lung accumulation was seen at day 3 in mice infected by inhalation with A. fumigatus. By day 8, coincident with increasing anti-ATAK antibodies, luminescence signal was lost and there was no detectable mRNA transcription from ATAK cells. CONCLUSIONS: ATAK cells accumulated in target organs with distinct profiles, depending on the microbial etiology of infection. Finally, generation of an anti-ATAK immune response may provide an important safety mechanism that helps clear the cells from the host as the marrow recovers.


Assuntos
Infecções por Acinetobacter/imunologia , Candidíase/imunologia , Células Matadoras Naturais/imunologia , Aspergilose Pulmonar/imunologia , RNA Mensageiro/metabolismo , Infecções Estafilocócicas/imunologia , Infecções por Acinetobacter/microbiologia , Infecções por Acinetobacter/terapia , Acinetobacter baumannii , Animais , Aspergillus fumigatus , Candida albicans , Candidíase/microbiologia , Candidíase/terapia , Ciclofosfamida , Gliceraldeído-3-Fosfato Desidrogenases/genética , Células HL-60 , Humanos , Imunoterapia Adotiva , Rim/imunologia , Luminescência , Pulmão/imunologia , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neutropenia/induzido quimicamente , Aspergilose Pulmonar/microbiologia , Aspergilose Pulmonar/terapia , Baço/imunologia , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/terapia , Staphylococcus aureus , Timidina Quinase/genética , Fatores de Tempo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA