Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
J Antimicrob Chemother ; 78(9): 2228-2241, 2023 09 05.
Article in English | MEDLINE | ID: mdl-37522316

ABSTRACT

BACKGROUND: Biofilms are the leading cause of nosocomial infections and are hard to eradicate due to their inherent antimicrobial resistance. Candida albicans is the leading cause of nosocomial fungal infections and is frequently co-isolated with the bacterium Pseudomonas aeruginosa from biofilms in the cystic fibrosis lung and severe burn wounds. The presence of C. albicans in multispecies biofilms is associated with enhanced antibacterial resistance, which is largely mediated through fungal extracellular carbohydrates sequestering the antibiotics. However, significantly less is known regarding the impact of polymicrobial biofilms on antifungal resistance. RESULTS: Here we show that, in dual-species biofilms, P. aeruginosa enhances the susceptibility of C. albicans to amphotericin B, an effect that was biofilm specific. Transcriptional analysis combined with gene ontology enrichment analysis identified several C. albicans processes associated with oxidative stress to be differentially regulated in dual-species biofilms, suggesting that P. aeruginosa exerts oxidative stress on C. albicans, likely through the secretion of phenazines. However, the mitochondrial superoxide dismutase SOD2 was significantly down-regulated in the presence of P. aeruginosa. Monospecies biofilms of the sod2Δ mutant were more susceptible to amphotericin B, and the susceptibility of these biofilms was further enhanced by exogenous phenazines. CONCLUSIONS: We propose that in dual-species biofilms, P. aeruginosa simultaneously induces mitochondrial oxidative stress, while down-regulating key detoxification enzymes, which prevents C. albicans mounting an appropriate oxidative stress response to amphotericin B, leading to fungal cell death. This work highlights the importance of understanding the impact of polymicrobial interactions on antimicrobial susceptibility.


Subject(s)
Amphotericin B , Candida albicans , Amphotericin B/pharmacology , Pseudomonas aeruginosa , Biofilms , Anti-Bacterial Agents/pharmacology , Phenazines , Antifungal Agents/pharmacology
2.
Microbiology (Reading) ; 169(1)2023 01.
Article in English | MEDLINE | ID: mdl-36748572

ABSTRACT

Pseudomonas aeruginosa is a Gram-negative opportunistic pathogen frequently isolated from chronic infections of the cystic fibrosis lung and burn wounds, and is a major cause of antimicrobial-resistant nosocomial infections. P. aeruginosa is frequently co-isolated with the opportunistic fungal pathogen Candida albicans, with the presence of C. albicans in dual-species biofilms promoting tolerance to meropenem. Here, transcription profiling of mature P. aeruginosa single- or dual-species biofilms was carried out to understand the molecular mechanism(s) by which C. albicans enhances meropenem tolerance. C. albicans appeared to have a mild impact on the transcriptome of P. aeruginosa mature biofilms, with most differentially regulated genes being involved in interkingdom interactions (i.e. quorum sensing and phenazine biosynthesis). The addition of meropenem to mature single- or dual-species biofilms resulted in a significant bacterial transcriptional response, including the induction of the beta-lactamase, ampC, genes involved in biofilm formation. P. aeruginosa elicited a similar transcriptional response to meropenem in the presence of C. albicans, but C. albicans promoted the expression of additional efflux pumps, which could play roles in increasing the tolerance of P. aeruginosa to meropenem.


Subject(s)
Biofilms , Pseudomonas aeruginosa , Pseudomonas aeruginosa/genetics , Meropenem/pharmacology , Candida albicans/physiology , Quorum Sensing/genetics
3.
Methods Mol Biol ; 2542: 309-321, 2022.
Article in English | MEDLINE | ID: mdl-36008675

ABSTRACT

The cell wall of Candida albicans is a multilayered structure consisting of polysaccharides and proteins. The inner cell wall layer is comprised of chitin and ß1-3 and ß1-6-glucan which contribute to the overall shape and structure of the cell, while the outer layer of highly glycosylated mannoproteins provides key functional traits such as cell adhesion required for virulence. However, the cell wall is not a static structure but is constantly being remodeled in response to the external environment. Given that all of the cell wall components act as pathogen-associated molecular patterns (PAMPs) that are recognized by a variety of receptors on the surface of innate immune cells, remodeling of the cell wall can have a dramatic impact on the host-pathogen interaction. For example, during growth in standard media, C. albicans shields its major cell wall PAMPs from the innate immune system, but during growth in acidic environments as encountered during colonization of the female reproductive tract, key PAMPs become exposed on the fungal cell surface initiating a strong pro-inflammatory innate immune response. The impact of environmental adaptation on fungal cell wall remodeling, and the subsequent impact this has on the host-pathogen interaction, has been the subject of much research. In this chapter, we outline techniques to assess cell wall components in both resting and environmentally adapted C. albicans cells.


Subject(s)
Candida albicans , Pathogen-Associated Molecular Pattern Molecules , Cell Wall/chemistry , Host-Pathogen Interactions , Immunity, Innate , Pathogen-Associated Molecular Pattern Molecules/metabolism
4.
Cell Surf ; 8: 100074, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35097244

ABSTRACT

Pathogenic fungi hide from their hosts by camouflage, obscuring immunogenic cell wall components such as beta-glucan with innocuous coverings such as mannoproteins and alpha-glucan that are less readily recognised by the host. Attempts to understand how such processes are regulated have met with varying success. Typically studies focus on understanding the transcriptional response of fungi to either their reservoir environment or the host. However, such approaches do not fully address this research question, due to the layers of post-transcriptional and post-translational regulation that occur within a cell. Although in animals the impact of post-transcriptional and post-translational regulation has been well characterised, our knowledge of these processes in the fungal kingdom is more limited. Mutations in RNA-binding proteins, like Ssd1 and Candida albicans Slr1, affect cell wall composition and fungal virulence indicating that post-transcriptional regulation plays a key role in these processes. Here, we review the current state of knowledge of fungal post-transcriptional regulation, and link this to potential mechanisms of immune evasion by drawing on studies from model yeast and plant pathogenic fungi. We highlight several RNA-binding proteins that regulate cell wall synthesis and could be involved in local translation of cell wall components. Expanding our knowledge on post-transcriptional regulation in human fungal pathogens is essential to fully comprehend fungal virulence strategies and for the design of novel antifungal therapies.

6.
Cell Rep ; 38(1): 110183, 2022 01 04.
Article in English | MEDLINE | ID: mdl-34986357

ABSTRACT

Candida albicans is a commensal of the urogenital tract and the predominant cause of vulvovaginal candidiasis (VVC). Factors that increase circulatory estrogen levels such as pregnancy, the use of oral contraceptives, and hormone replacement therapy predispose women to VVC, but the reasons for this are largely unknown. Here, we investigate how adaptation of C. albicans to estrogen impacts the fungal host-pathogen interaction. Estrogen promotes fungal virulence by enabling C. albicans to avoid the actions of the innate immune system. Estrogen-induced innate immune evasion is mediated via inhibition of opsonophagocytosis through enhanced acquisition of the human complement regulatory protein, Factor H, on the fungal cell surface. Estrogen-induced accumulation of Factor H is dependent on the fungal cell surface protein Gpd2. The discovery of this hormone-sensing pathway might pave the way in explaining gender biases associated with fungal infections and may provide an alternative approach to improving women's health.


Subject(s)
Candida albicans/immunology , Candidiasis, Vulvovaginal/pathology , Complement Pathway, Alternative/immunology , Estrogens/metabolism , Immune Evasion/immunology , Phagocytosis/immunology , Candida albicans/pathogenicity , Complement Factor H/metabolism , Female , Glycerol-3-Phosphate Dehydrogenase (NAD+)/metabolism , Humans , Immunity, Innate/immunology , Progesterone/metabolism , Virulence/immunology
7.
J Fungi (Basel) ; 7(2)2021 Feb 06.
Article in English | MEDLINE | ID: mdl-33562068

ABSTRACT

Candida albicans infections range from superficial to systemic and are one of the leading causes of fungus-associated nosocomial infections. The innate immune responses during these various infection types differ, suggesting that the host environment plays a key role in modulating the host-pathogen interaction. In addition, C. albicans is able to remodel its cell wall in response to environmental conditions to evade host clearance mechanisms and establish infection in niches, such as the oral and vaginal mucosa. Phagocytes play a key role in clearing C. albicans, which is primarily mediated by Pathogen Associated Molecular Pattern (PAMP)-Pattern Recognition Receptor (PRR) interactions. PRRs such as Dectin-1, DC-SIGN, and TLR2 and TLR4 interact with PAMPs such as ß-glucans, N-mannan and O-mannan, respectively, to trigger the activation of innate immune cells. Innate immune cells exhibit distinct yet overlapping repertoires of PAMPs, resulting in the preferential recognition of particular Candida morphotypes by them. The role of phagocytes in the context of individual infection types also differs, with neutrophils playing a prominent role in kidney infections, and dendritic cells playing a prominent role in skin infections. In this review, we provide an overview of the key receptors involved in the detection of C. albicans and discuss the differential innate immune responses to C. albicans seen in different infection types such as vulvovaginal candidiasis (VVC) and oral candidiasis.

8.
J Antimicrob Chemother ; 75(4): 925-935, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31865379

ABSTRACT

BACKGROUND: Pseudomonas aeruginosa is an opportunistic bacterium that infects the airways of cystic fibrosis patients, surfaces of surgical and burn wounds, and indwelling medical devices. Patients are prone to secondary fungal infections, with Candida albicans being commonly co-isolated with P. aeruginosa. Both P. aeruginosa and C. albicans are able to form extensive biofilms on the surfaces of mucosa and medical devices. OBJECTIVES: To determine whether the presence of C. albicans enhances antibiotic tolerance of P. aeruginosa in a dual-species biofilm. METHODS: Single- and dual-species biofilms were established in microtitre plates and the survival of each species was measured following treatment with clinically relevant antibiotics. Scanning electron microscopy and confocal microscopy were used to visualize biofilm structure. RESULTS: C. albicans enhances P. aeruginosa biofilm tolerance to meropenem at the clinically relevant concentration of 5 mg/L. This effect is specific to biofilm cultures and is dependent upon C. albicans extracellular matrix polysaccharides, mannan and glucan, with C. albicans cells deficient in glycosylation structures not enhancing P. aeruginosa tolerance to meropenem. CONCLUSIONS: We propose that fungal mannan and glucan secreted into the extracellular matrix of P. aeruginosa/C. albicans dual-species biofilms play a central role in enhancing P. aeruginosa tolerance to meropenem, which has direct implications for the treatment of coinfected patients.


Subject(s)
Candida albicans , Pseudomonas aeruginosa , Biofilms , Drug Tolerance , Humans , Meropenem/pharmacology
9.
mBio ; 10(5)2019 10 15.
Article in English | MEDLINE | ID: mdl-31615961

ABSTRACT

Candida albicans is a commensal yeast of the human gut which is tolerated by the immune system but has the potential to become an opportunistic pathogen. One way in which C. albicans achieves this duality is through concealing or exposing cell wall pathogen-associated molecular patterns (PAMPs) in response to host-derived environment cues (pH, hypoxia, and lactate). This cell wall remodeling allows C. albicans to evade or hyperactivate the host's innate immune responses, leading to disease. Previously, we showed that adaptation of C. albicans to acidic environments, conditions encountered during colonization of the female reproductive tract, induces significant cell wall remodeling resulting in the exposure of two key fungal PAMPs (ß-glucan and chitin). Here, we report that this pH-dependent cell wall remodeling is time dependent, with the initial change in pH driving cell wall unmasking, which is then remasked at later time points. Remasking of ß-glucan was mediated via the cell density-dependent fungal quorum sensing molecule farnesol, while chitin remasking was mediated via a small, heat-stable, nonproteinaceous secreted molecule(s). Transcript profiling identified a core set of 42 genes significantly regulated by pH over time and identified the transcription factor Efg1 as a regulator of chitin exposure through regulation of CHT2 This dynamic cell wall remodeling influenced innate immune recognition of C. albicans, suggesting that during infection, C. albicans can manipulate the host innate immune responses.IMPORTANCECandida albicans is part of the microbiota of the skin and gastrointestinal and reproductive tracts of humans and has coevolved with us for millennia. During that period, C. albicans has developed strategies to modulate the host's innate immune responses, by regulating the exposure of key epitopes on the fungal cell surface. Here, we report that exposing C. albicans to an acidic environment, similar to the one of the stomach or vagina, increases the detection of the yeast by macrophages. However, this effect is transitory, as C. albicans is able to remask these epitopes (glucan and chitin). We found that glucan remasking is controlled by the production of farnesol, a molecule secreted by C. albicans in response to high cell densities. However, chitin-remasking mechanisms remain to be identified. By understanding the relationship between environmental sensing and modulation of the host-pathogen interaction, new opportunities for the development of innovative antifungal strategies are possible.


Subject(s)
Antifungal Agents/pharmacology , Candida albicans/drug effects , Candida albicans/physiology , Quorum Sensing/genetics , beta-Glucans/metabolism , Candida albicans/genetics , Cell Wall/metabolism , Chitin/metabolism , Glucans/metabolism , Hydrogen-Ion Concentration
10.
Sci Rep ; 9(1): 5714, 2019 04 05.
Article in English | MEDLINE | ID: mdl-30952923

ABSTRACT

Rhizopus spp are the most common etiological agents of mucormycosis, causing over 90% mortality in disseminated infection. Key to pathogenesis is the ability of fungal spores to swell, germinate, and penetrate surrounding tissues. Antibiotic treatment in at-risk patients increases the probability of the patient developing mucormycosis, suggesting that bacteria have the potential to control the growth of the fungus. However, research into polymicrobial relationships involving Rhizopus spp has not been extensively explored. Here we show that co-culturing Rhizopus microsporus and Pseudomonas aeruginosa results in the inhibition of spore germination. This inhibition was mediated via the secretion of bacterial siderophores, which induced iron stress on the fungus. Addition of P. aeruginosa siderophores to R. microsporus spores in the zebrafish larval model of infection resulted in inhibition of fungal germination and reduced host mortality. Therefore, during infection antibacterial treatment may relieve bacterial imposed nutrient restriction resulting in secondary fungal infections.


Subject(s)
Iron/metabolism , Microbial Interactions , Pseudomonas aeruginosa/physiology , Rhizopus/growth & development , Siderophores/metabolism , Zebrafish/microbiology , Animals , Antifungal Agents , Female , Male , Mucormycosis , Pseudomonas Infections , Pseudomonas aeruginosa/metabolism
11.
Article in English | MEDLINE | ID: mdl-32047726

ABSTRACT

Due to limited mobility, fungi, like most unicellular organisms, have evolved mechanisms to adapt to sudden chemical and/or physical variation in their environment. Candida albicans is recognized as a model organism to study eukaryotic responses to environmental changes, as this human commensal yeast but also opportunistic pathogen responds to numerous environmental cues through switching morphologies from yeast to hyphae growth. This mechanism is largely controlled by two major pathways: cAMP-PKA and MAPK, but each environmental signal is sensed by specific sensors. However, morphological switching is not the only response C. albicans exerts in response to environmental cues. Recently, fungal cell wall remodeling in response to host-derived environmental cues has been identified as a way for C. albicans to manipulate the innate immune system. The fungal cell wall is composed of a chitin skeleton linked to a network of ß-glucan, which anchors proteins and mannans to the fungal cell surface. As localized on the cell surface, these molecules drive interactions with the environment and other cells, particularly with host immune cells. C. albicans is recognized by immune cells such as neutrophils and macrophages via pathogen recognition receptors (PRRs) that bind different components of the cell wall. While ß-glucan and mannan are proinflammatory molecules, chitin can induce anti-inflammatory responses. Interestingly, C. albicans is able to regulate the exposure of these pathogen-associated molecular patterns (PAMPs) according to environmental cues resulting in a modulation of the host immune response. This review describes the mechanisms involved in C. albicans response to environmental changes and their effect on immune recognition.


Subject(s)
Candida albicans/immunology , Candida albicans/physiology , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/physiology , Immunity, Innate , Candida albicans/cytology , Cell Wall/metabolism , Chitin/metabolism , Humans , Macrophages , Mannans/metabolism , Neutrophils , beta-Glucans/metabolism
12.
Adv Appl Microbiol ; 102: 159-221, 2018.
Article in English | MEDLINE | ID: mdl-29680125

ABSTRACT

The ability to cause disease extends from the ability to grow within the host environment. The human host provides a dynamic environment to which fungal pathogens must adapt to in order to survive. The ability to grow under a particular condition (i.e., the ability to grow at mammalian body temperature) is considered a fitness attribute and is essential for growth within the human host. On the other hand, some environmental conditions activate signaling mechanisms resulting in the expression of virulence factors, which aid pathogenicity. Therefore, pathogenic fungi have evolved fitness and virulence attributes to enable them to colonize and infect humans. This review highlights how some of the major pathogenic fungi respond and adapt to key environmental signals within the human host.


Subject(s)
Adaptation, Physiological , Fungi/growth & development , Fungi/pathogenicity , Host-Pathogen Interactions , Mycoses/microbiology , Gene Expression Regulation, Fungal , Humans , Signal Transduction , Virulence Factors/biosynthesis
13.
J Immunol ; 200(10): 3539-3546, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29643192

ABSTRACT

The pathogenic fungus Cryptococcus enters the human host via inhalation into the lung and is able to reside in a niche environment that is serum- (opsonin) limiting. Little is known about the mechanism by which nonopsonic phagocytosis occurs via phagocytes in such situations. Using a combination of soluble inhibitors of phagocytic receptors and macrophages derived from knockout mice and human volunteers, we show that uptake of nonopsonized Cryptococcus neoformans and C. gattii via the mannose receptor is dependent on macrophage activation by cytokines. However, although uptake of C. neoformans is via both dectin-1 and dectin-2, C. gattii uptake occurs largely via dectin-1. Interestingly, dectin inhibitors also blocked phagocytosis of unopsonized Cryptococci in wax moth (Galleria mellonella) larvae and partially protected the larvae from infection by both fungi, supporting a key role for host phagocytes in augmenting early disease establishment. Finally, we demonstrated that internalization of nonopsonized Cryptococci is not accompanied by the nuclear translocation of NF-κB or its concomitant production of proinflammatory cytokines such as TNF-α. Thus, nonopsonized Cryptococci are recognized by mammalian phagocytes in a manner that minimizes proinflammatory cytokine production and potentially facilitates fungal pathogenesis.


Subject(s)
Cryptococcosis/metabolism , Cryptococcosis/microbiology , Cryptococcus gattii/pathogenicity , Cryptococcus neoformans/pathogenicity , Macrophages/metabolism , Macrophages/microbiology , Animals , Cell Line , Cytokines/metabolism , Humans , Lectins, C-Type/metabolism , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred C57BL , Moths , NF-kappa B/metabolism , Opsonin Proteins/metabolism , Phagocytes/metabolism , Phagocytes/microbiology , Phagocytosis/physiology , Receptors, Cell Surface/metabolism , Tumor Necrosis Factor-alpha/metabolism
14.
PLoS One ; 13(3): e0194615, 2018.
Article in English | MEDLINE | ID: mdl-29596441

ABSTRACT

Cryptococcosis remains the leading cause of fungal meningitis worldwide, caused primarily by the pathogen Cryptococcus neoformans. Symptomatic cryptococcal infections typically affect immunocompromised patients. However, environmental exposure to cryptococcal spores is ubiquitous and most healthy individuals are thought to harbor infections from early childhood onwards that are either resolved, or become latent. Since macrophages are a key host cell for cryptococcal infection, we sought to quantify the extent of individual variation in this early phagocyte response within a small cohort of healthy volunteers with no reported immunocompromising conditions. We show that rates of both intracellular fungal proliferation and non-lytic expulsion (vomocytosis) are remarkably variable between individuals. However, we demonstrate that neither gender, in vitro host inflammatory cytokine profiles, nor polymorphisms in several key immune genes are responsible for this variation. Thus the data we present serve to quantify the natural variation in macrophage responses to this important human pathogen and will hopefully provide a useful "benchmark" for the research community.


Subject(s)
Cryptococcus neoformans/physiology , Genetic Variation , Healthy Volunteers , Macrophages/microbiology , Environment , Humans
15.
Trends Microbiol ; 26(4): 284-295, 2018 04.
Article in English | MEDLINE | ID: mdl-29452950

ABSTRACT

Deadly infections from opportunistic fungi have risen in frequency, largely because of the at-risk immunocompromised population created by advances in modern medicine and the HIV/AIDS pandemic. This review focuses on dynamics of the fungal polysaccharide cell wall, which plays an outsized role in fungal pathogenesis and therapy because it acts as both an environmental barrier and as the major interface with the host immune system. Human fungal pathogens use architectural strategies to mask epitopes from the host and prevent immune surveillance, and recent work elucidates how biotic and abiotic stresses present during infection can either block or enhance masking. The signaling components implicated in regulating fungal immune recognition can teach us how cell wall dynamics are controlled, and represent potential targets for interventions designed to boost or dampen immunity.


Subject(s)
Adaptation, Physiological , Cell Wall/immunology , Fungal Proteins/immunology , Fungi/immunology , Immune Evasion , Mycoses/immunology , Cell Wall/chemistry , Epitopes , Fungi/chemistry , Fungi/pathogenicity , Glucans/immunology , Host-Pathogen Interactions/immunology , Humans , Hydrogen-Ion Concentration , Immunity, Innate , Lactic Acid , Polysaccharides
16.
Curr Opin Microbiol ; 40: 58-64, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29132066

ABSTRACT

Fungi are ubiquitous transient or persistent human colonisers, and form the mycobiome with shifts in niche specific mycobiomes (dysbiosis) being associated with various diseases. These complex interactions of fungal species with the human host can be viewed as a spectrum of symbiotic relationships (i.e. commensal, parasitic, mutualistic, amensalistic). The host relevant outcome of the relationship is the damage to benefit ratio, elegantly described in the damage response framework. This review focuses on Candida albicans, which is the most well studied human fungal symbiont clinically and experimentally, its transition from commensalism to parasitism within the human host, and the factors that influence this relationship.


Subject(s)
Candida albicans/physiology , Candidiasis/microbiology , Candidiasis/physiopathology , Symbiosis , Animals , Candida albicans/genetics , Host-Pathogen Interactions , Humans
18.
PLoS Pathog ; 13(5): e1006403, 2017 May.
Article in English | MEDLINE | ID: mdl-28542528

ABSTRACT

Candida albicans is able to proliferate in environments that vary dramatically in ambient pH, a trait required for colonising niches such as the stomach, vaginal mucosal and the GI tract. Here we show that growth in acidic environments involves cell wall remodelling which results in enhanced chitin and ß-glucan exposure at the cell wall periphery. Unmasking of the underlying immuno-stimulatory ß-glucan in acidic environments enhanced innate immune recognition of C. albicans by macrophages and neutrophils, and induced a stronger proinflammatory cytokine response, driven through the C-type lectin-like receptor, Dectin-1. This enhanced inflammatory response resulted in significant recruitment of neutrophils in an intraperitoneal model of infection, a hallmark of symptomatic vaginal colonisation. Enhanced chitin exposure resulted from reduced expression of the cell wall chitinase Cht2, via a Bcr1-Rim101 dependent signalling cascade, while increased ß-glucan exposure was regulated via a non-canonical signalling pathway. We propose that this "unmasking" of the cell wall may induce non-protective hyper activation of the immune system during growth in acidic niches, and may attribute to symptomatic vaginal infection.


Subject(s)
Candida albicans/immunology , Candidiasis/immunology , Cell Wall/immunology , Animals , Candida albicans/physiology , Candidiasis/microbiology , Cell Wall/chemistry , Humans , Hydrogen-Ion Concentration , Immunity, Innate , Macrophages/immunology , Macrophages/microbiology , Mice
19.
J Fungi (Basel) ; 3(4)2017 Oct 07.
Article in English | MEDLINE | ID: mdl-29371572

ABSTRACT

With the advent of high-throughput sequencing techniques, the astonishing extent and complexity of the microbial communities that reside within and upon us has begun to become clear. Moreover, with advances in computing and modelling methods, we are now beginning to grasp just how dynamic our interactions with these communities are. The diversity of both these communities and their interactions-both within the community and with us-are dependent on a multitude of factors, both microbial- and host-mediated. Importantly, it is becoming clear that shifts in the makeup of these communities, or their responses, are linked to different disease states. Although much of the work to define these interactions and links has been investigating bacterial communities, recently there has been significant growth in the body of knowledge, indicating that shifts in the host fungal communities (mycobiome) are also intimately linked to disease status. In this review, we will explore these associations, along with the interactions between fungal communities and their human and microbial habitat, and discuss the future applications of systems biology in determining their role in disease status.

20.
Int J Antimicrob Agents ; 48(1): 69-77, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27289450

ABSTRACT

The fungal pathogen Cryptococcus neoformans poses a major threat to immunocompromised patients and is a leading killer of human immunodeficiency virus (HIV)-infected patients worldwide. Cryptococci are known to manipulate host macrophages and can either remain latent or proliferate intracellularly within the host phagocyte, a favourable niche that also renders them relatively insensitive to antifungal agents. Here we report an attempt to address this limitation by using a fluorescence-based drug screening method to identify potential inhibitors of intracellular proliferation of C. neoformans. The Prestwick Chemical Library(®) of FDA-approved small molecules was screened for compounds that limit the intracellular replication of a fluorescently-tagged C. neoformans reference strain (H99-GFP) in macrophages. Preliminary screening revealed 19 of 1200 compounds that could significantly reduce intracellular growth of the pathogen. Secondary screening and host cell cytotoxicity assays highlighted fendiline hydrochloride as a potential drug candidate for the development of future anticryptococcal therapies. Live cell imaging demonstrated that this Ca(2+) channel blocker strongly enhanced phagosome maturation in macrophages leading to improved fungal killing and reduced intracellular replication. Whilst the relatively high dose of fendiline hydrochloride required renders it unfit for clinical deployment against cryptococcosis, this study highlights a novel approach for identifying new lead compounds and unravels a pharmacologically promising scaffold towards the development of novel antifungal therapies for this neglected disease.


Subject(s)
Antifungal Agents/pharmacology , Cryptococcus neoformans/drug effects , Cytological Techniques/methods , Drug Evaluation, Preclinical/methods , Macrophages/microbiology , Animals , Cell Line , Cell Survival/drug effects , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...