Open access peer-reviewed chapter

Pathogenicity Mechanism of Candida albicans

Written By

Snigdha Pattnaik, Laxmidhar Maharana and Manoj Sethi

Submitted: 17 November 2020 Reviewed: 30 July 2021 Published: 05 September 2021

DOI: 10.5772/intechopen.99737

From the Edited Volume

Advances in Candida albicans

Edited by Xinhui Wang

Chapter metrics overview

787 Chapter Downloads

View Full Metrics

Abstract

In normal human microbiome, the polymorphic fungus Candida albicans is a crucial member. C. albicans resides mostly in individual as harmless commensal life. In specific situations, however, C. albicans can cause diseases that cause contaminations of the skin to life-threatening fundamental contaminations. Pathogenesis of Candida species is contributed by multiple factors. Some of the major contributors are enlisted here. These include host pathogen interaction, receptors molecule like TLR recognition, TLR signaling, C type lectin receptors, Dectin 1,2 and 3, mannose receptor, mincle, DC sign, Nod-Like Receptors (NLRs) and inflammasomes, soluble molecules in candida recognition, cellular responses to candida such as neutrophils, macrophages. This chapter enlightens all the components of candida pathogenicity by the assessment of Candida species pathogenic determinants. All together these will explain the current knowledge about how these determinant factors and receptors modulate virulence as well as consequent infection. Better understanding of candida pathogenicity mechanism can be the resultant of better treatment guidelines along with development of novel antifungal agents. Overall, in this review we present an update in the current understanding of the insight of pathogenicity mechanisms in this important human pathogen.

Keywords

  • Pathogenicity
  • C. albicans
  • TLR
  • receptor
  • lectin

1. Introduction

Candida is a diploid parasite that as often as possible causes mucosal and fundamental contaminations in people [1]. Candida species can colonize a few particular anatomical locales. Greater part of diseases by commensal microorganisms comes from endogenous colonization. Notwithstanding, exogenous pollution, for example, diseases communicated through emergency clinic workers, medical clinic air, and biofilm-debased intrusive gadgets like catheters, can likewise happen [2, 3, 4]. Diseases brought about by Candida can be delegated shallow, cutaneous, mucosal, and fundamental infection. At the point when Candida spp. taint the oral cavity, skin, genitalia, respiratory framework, and the remainder of the gastrointestinal lot, the disease is delegated the shallow sort. Intrusive candidiasis is a disease portrayed with very extreme conditions, for example, candidemia, meningitis (influencing the mind), and endocarditis (influencing the heart) [5]. In hospitalized patients and those with bedraggled safe framework, intrusive contamination is a huge reason for dismalness and mortality along with increased frequency as well as pervasiveness rates.

Candida species pathogenesis is a complex cycle including numerous instruments and pathways. It is likewise a mind boggling and multifactorial system, including highlights of both the host and the microorganism [6]. For contamination to be set up, the pioneering microorganism should avoid, duplicate in the host climate, and make do in the safe arrangement of the host. The living being must likewise have the option to scatter to other body tissues and organs, most particularly in foundational disease [7]. Problem in skin or gastrointestinal boundaries can prompt dispersed or profound organ candidiasis. In more significant circumstances, circulatory system intrusion may some time possible which hence will disperse to various organs of the body.

Candida contaminations in a great many people are asymptomatic. This is because of the capacity of the immunological framework to checkmate the life form as it endeavors to spread in the body. In any case, consumption in resistant systemor changes in microbiota balance, combined with different elements, can work with the spread of Candida which is regularly deadly in 42% of announced cases [8, 9, 10]. C. albicans is answerable for about half of candidiasis and non-albicans Candida species are liable for the rest of the Candida contamination. Disease brought about by several other species of candida are of extraordinary concern. A portion of these non-albicans Candida species are presently viewed as arising artful microbes [11]. Forestalling Candida contaminations for the most part brought about by Candida species is a developing test in human medication. Indeed, even with the accessibility and utilization of antifungal prescription, scattered candidiasis is went with high death rate (around 40–60%), helpless conclusion, and unseemly illness the board. The overall clinical show of the patient likewise adds to the expansion in death rate. Protection from antifungal medications is not, at this point another issue. Indeed, even among people that have not been presented to anti-infection agents, obstruction has been accounted for [12]. Candida is one of the main sources of mucosal contaminations in sound people for now days. It additionally causes initial diseases particularly in immunosuppressive patients, regardless of its status as a commensal microorganism [13]. Truth be told, candidiasis is viewed as the third to fourth most regular infection in medical care offices inside the USA and even all around the world [14].

As anyone might expect, it is the destructiveness and pathogenic qualities and components that have gotten the most consideration from specialists throughout the long term. As of late, much have been found out about the components of Candida pathogenesis. Studies have shown that at the core of the capacity of Candida to multiply, change from non-destructiveness commensal to pioneering pathogenic organism and build up disease in the host lie profoundly interconnected elements made out of transcriptional circuits, morphology-related/harmfulness encoding qualities, metabolic versatility, genome pliancy, phenotypic exchanging, biofilm arrangement, tissue harming extracellular hydrolytic catalysts, and a few different variables that work with destructiveness and pathogenesis in Candida species [15]. Changes in ecological pH, vigorous supplement procurement framework, escape from phagocytosis, avoidance from have insusceptible framework, have microbiome coaggregation, protection from antifungal specialists, and the capacity to productively react to numerous anxieties are other crucial characteristics that upgrade endurance and pathogenesis.

In order to be capable of inducing such a diversity of infections C. albicans can live in several anatomically discrete sites and translates several virulence factors. The phenomenon of phenotypic converting from yeast- to filament-growth is just one, but critical, factor that contributes to the virulence of C. albicans. It offers a basis for activating different receptors leading to diverse immune responses. Other virulence factors of C. albicans contain adhesion factors, thigmotropism and secretion of several hydrolytic enzymes, such as lipase, phospholipase, and proteinase. During the past few years it has become increasingly clear that PRRs are vital for the host response to C. albicans, with various TLRs and LRs having distinctive roles in innate immunity. Each ligand–receptor system activates specific intracellular signaling pathways, which in turn leads to modulation of various components of the host immune response. While a few receptors, like TLR4, dectin 1 and the MR, apply an all the more favorable to fiery job, others employ immunosuppressive impacts (for instance, TLR2, CR3 and FcγR). After disclosure and characterized clarification of the part of TLRs in parasitic acknowledgment, further investigations have explained the job of the C-type lectin receptors with an emphasis basically on dectin-1 and dectin-2. The presence of various relationships among all of the components that guide the establishment of pollutions is an undeniable component in the pathogenesis of Candida species. This chapter is precisely based on the mechanisms of Candida pathogenesis with emphasis on the virulence factors mostly the important receptors and pathogenic determinants.

Advertisement

2. Pathogenicity mechanism of Candida species

2.1 Infection

The pathogenicity of C. albicans is identified with its change between the commensal yeast structure and the obtrusive hyphal shape [16]. Upon have cell connection, thigmotropism (contact detecting) triggers C. albicans filamentation. This allows the creature to infiltrate further into the host tissues through extracellular compound emission [17]. The capacity of Candida to change over from yeast to hyphae stage or hyphae to yeast stage is named dimorphism. Every one of these periods of development is crucial for harmfulness and pathogenicity as it impacts how Candida gets away from the resistant framework. Yeast and fiber (hyphae) structures assume autonomous parts during scattered candidiasis. While the yeast structures engaged with scattering, the hyphal (filamentous) structure is associated with tissue intrusion and pathogenesis [18]. Candida species should have the option to adequately colonize its host and moreover adjust to assortments of unessential requirements like temperature, oxygen, pH, carbon dioxide, and diverse negative organic conditions, for example, carbon source, supplement accessibility, the immunological framework, and other existing together bacterial and contagious cells inside the specialty [19, 20]. Positive reaction to those imperatives has a quick impact in transformation and advancement of Candida harmfulness and pathogenicity. Before receptor-intervened epithelial acknowledgment by Candida species, a few flagging pathways are actuated. Temperature change, supplement starvation, oxidative pressure, osmotic pressure, and pH detecting trigger mitogen-enacted protein kinase, pathways based on CAMP, transduction of Rim-101, along with surprisingly hereditary mechanisms that constantly instigate numerous qualities. Most of the induced characteristics are connected with filamentous turn of events and biofilm plan. While assorted hereditary pathways transduce shifts from yeast to hyphae or hyphae to yeast stage, distinctive ecological signals emphatically and contrarily regulate morphology-related cell surface exchanging [21]. The flagging and variation pathways assume pivotal parts in different physiological and cell measures engaged with the Candida species pathogenicity as demonstrated in Table 1.

S/no.PathwaysFunctionsReference(s)
1Mitogen-activated protein kinase (MAPK) pathwaysImportant regulator of morphogenesis.[22, 23]
Involved in sensing and transmitting stress signals and other environmental signals
Three main MAP kinase pathways are the following:
  1. Mkc1- controls cellular integrity, invasive growth, cell wall biogenesis, and formation of biofilm

  2. Hog1- mediates response to thermal, osmotic, and oxidative stress. Controls cell wall formationand morphogenesis. Under osmotic stress, its activation leads to glycerol accumulation.

  3. Cek1- it mediates mating and hyphae formation and is also involved in adaptation to boththermal and nutrient stresses.

[24].
2Ras-CAMP-PKA pathwaysRegulate adhesion, dimorphism. Also involved in the formation of biofilms.[23, 25, 26]
Control hyphal formation and white-to-opaque change[27, 28]
Involved in drug tolerance and in the maintenance of cell wall integrity
3RIM 101 signal transductionEnables Candida albicans to sense pH changes, thus mediate pH-dependent responses[29]
4Stress response pathwaysContribute to virulence and pathogenesis
Facilitate adaptation to ever-changing environmental conditions.
Protect against host-derived stresses
[30]
5Ergosterol biosynthetic pathwaysLink between hyphae formation and virulence in C. albicans[31]
Enhance cell adhesion and damage to the tissues[32]
ERG3 and ERG11 play major roles in azole drug resistance; thus, it is the target of fluconazoleantifungals
6Genome plasticityTriggers adaptation to fluctuating host environment.
Leads to the generation of recombinant progeny with increased fitness.
Induces natural mutations that alter the balance between commensalism and pathogenicity.
[33, 34]
Facilitates resistance to stressors including antifungal agents and pathogenicity during systemic and mucosal infections[35]
Triggers polarized filamentous growth
Involved in the generation/evolution of new pathotypes or strains
Enhances the utilization of several nutrients.
Facilitates Candida growth rate, as well as its morphology and behaviors at the host interface
7Calcium-calcineurin pathwaysMajor mediator of stress responses[36]
Essential for survival in the presence of stressors[37]
Play crucial roles in virulence[38, 39]

Table 1.

Major pathogenicity inducing pathways/responses in Candida species.

The greater part of the flagging pathways are amazingly fundamental for protecting Candida spp. against immunological assault [40]. They assume different parts in the declaration of morphology related qualities. The co-articulation of morphology-connected proteins brings about synergistic association among quality items fundamental for biofilm foundation and development inside the host [41]. Along these lines, for hindering Candida endurance in have tissues, impedance with Candida species capacity to incorporate quality articulation to changes in morphology could be surely a potential restorative technique [42]. Also, distinguishing flagging segments saved among Candida species is vital for recognizing potential medication targets. During the interaction of pathogenesis, actuated endocytosis happens. It for the most part happens inside 4 h of starting contact to epithelial cell. Candida uses prompted endocytosis to sidestep invulnerable acknowledgment. The acknowledgment of invasins communicated on the contagious cell surface triggers prompted endocytosis. Until this point, only A1s3p and Ssa1p (invasins) are known for C. albicans. In a murine model of oropharyngeal candidiasis revealed by Sun et al., Als3 and Ssa1 freaks displayed diminished grip and intrusion of cells of epithelium [43]. Free of the cellular receptor of epithelium, instigated endocytosis can likewise happen. This is conceivable through the association of the host epithelial cell epidermal development factor receptor with the invasins of candida cell. Post actuated endocytosis, discharged harmfulness factors by pathogens to improve capacity to enter to surface of mucosa. The oral and vaginal mucosa, which are terminally separated and non-proliferative, are made out of delineated layers more averse to work with intrusion of parasites by means of initiated endocytosis. Candida species should use an elective course to attack a tissue less inclined to help disguise in a cycle called dynamic infiltration. Dynamic infiltration interceded through hyphae augmentation (constrained by Ume6 and Eed1) is a contagious actuated cycle that needs reasonable parasitic hyphae [44]. Actual powers, attachment, and hydrolytic chemicals like SAP additionally assume a part. C. albicans uses dynamic entrance as the underlying way to attack the furthest layers of the epithelium in vivo. Be that as it may, prompted endocytosis could likewise be obvious of additional upgraded attack once the fundamental proliferative layers of the epithelium have been gotten to by the growth. Along these lines, both dynamic infiltration and initiated endocytosis are unthinkingly noticeable systems required for disease foundation through mucosal boundaries in vivo. When all is said in done, the pathogenesis of Candida begins with colonization, shallow disease, and profound situated contamination before spread contamination. The overall strides in tissue intrusion by C. albicans incorporate in the following stages.

  1. Adhesion to the cellular epithelium.

  2. Colonization.

  3. Penetration to epithelium/hyphal invasion.

  4. Dissemination of vasculature.

  5. Endothelial colonization/penetration.

Systemic candida infection only occurs by immune system escape than vasculature penetration and invading the blood components. Entry to the bloodstream occurs via two routes:

  1. Natural routes.

  2. Artificial routes.

Above subsequent course is worked with biofilm arrangement as pathogens can get away and invade the blood. For Candida to endure and spread in the blood, various qualities are upregulated: qualities engaged with protein amalgamation, glycolytic cycle, glycolysis, and reaction to oxidative pressure. The presence of Candida in the blood prompts a condition called candidemia. From the blood, the yeast is dispersed to different fundamental organs in the body where it causes foundational contaminations. Dispersed candidiasis is profoundly worked with by extracellular hydrolytic compounds, adhesins, phenotypic exchanging, and cytolytic proteins. Candida in the blood can likewise bring about candiduria by antegrade contamination. Albeit most diseases include biofilm arrangement, a few contaminations can happen without the development of biofilm. Indeed, hyphae development and development are the beginning stages in the pathogenicity of Candida species, with the exception of C. glabrata that does not shape hyphae. It is notable that few qualities straightforwardly or by implication incited by natural irritations trigger hyphae arrangement.

Notwithstanding, questions actually remain with respect to the instruments controlling its union, the receptors, and its carrier. In outline, the exchanging of Candida spp. from commensal to artful microbe is ascribed to destructiveness factors that are specifically communicated under reasonable inclining conditions. The majority of these destructiveness factors are under close guideline. More examinations in their administrative instruments could be fundamental in the mission for new antifungal specialists. Figure 1 is the significant organization of Candida destructiveness and pathogenesis showing the associations between the different pathogenic determinants and harmful variables.

Figure 1.

Simplified diagram illustrating the network of Candida virulence and pathogenicity. (1) planktonic yeast cells attach to surfaces. Favorable conditions facilitate overgrowth; adherence (2): The cells attach to host cells via adhesins; hyphae formation/extension (3): Environmental constrains induce the HSPs, signaling and adaptation pathways which induce morphology-associated genes. The formation of the hyphae marks the beginning of Candida pathogenesis. Epithelial/endothelial adhesion/invasion (4 and 6): This is facilitated by hydrolytic enzymes and it is achieved via two ways: Induced endocytosis and active penetration. Some species such as C. glabrata do not form hyphae; rather, they form biofilms (5) prior to the establishment of infection. Destruction of epithelial and mucosal surfaces by the enzymes and cytolytic proteins gives rise to different types of candidiasis (8). Yeast cells can enter the blood (7) and then disseminate to the vital organs where they establish new biofilms. Infections associated with biofilms are of great clinical significance. Major Candida infections include vulvovaginal, oropharyngeal, and gastrointestinal candidiasis, candidemia, candiduria, and intra-abdominal candidiasis. Key: Dashed lines: Signals and inductions; single-headed thick dark red arrow: Major route of Candida pathogenesis; curved double-arrow connector: Interaction/association between factors; T-shaped thin red line: Inhibitory signal. The pool of virulence encoding genes house both the genes involved in hyphae and biofilm formation and other vital processes crucial for pathogenesis.

2.2 Host response to Candida species

Host insusceptible acknowledgment of Candida happens through a few instruments involving intrinsic and versatile insusceptibility. The versatile insusceptible framework perceives explicit antigenic moieties, prompting the advancement of a focused on safe reaction. Interestingly, inborn insusceptible acknowledgment is vague and wide and is the primary line of host protection against possibly hazardous organisms. These vague reactions are promptly endless supply of an organism in a pre-modified design and assume a fundamental part in controlling contagious weights and forestalling infection. Natural invulnerability includes a progression of dissolvable (supplement) and cell (neutrophil, macrophage) parts that act in show to keep by far most of microbes from setting up an intrusive disease. Further, it has become progressively clear that these reactions capacity to enact versatile insusceptibility just as acting along with other homeostatic cycles to give further security. Natural invulnerable acknowledgment of Candida happens through the acknowledgment of microorganism related atomic examples (PAMPs).

PAMPs are themes or particles that are regular between various sorts of growths. In contrast to antigens, individual PAMPs are not explicit to a solitary Candida animal variety but instead are divided among various species and contagious genera. These microbial PAMPs are perceived by have germline encoded design acknowledgment receptors (PRRs) [45] and give a pre-customized method of parasitic acknowledgment, taking into consideration moment acknowledgment of normal contagious parts. Most of contagious PAMPs are cell divider related and incorporate 𝛽-glucans, 𝑁-and 𝑂-connected mannans, and phospholipomannans [46]. These are perceived by three key PRR families: cost like receptors (TLRs), C-type lectin receptors (CLRs), and nucleotide-restricting area leucine-rich receptors (NLRs) [46, 47, 48, 49, 50, 51, 52]. Dendritic cells, monocytes, macrophages, polymorphonuclear leukocytes (PMNs), Tcells, Bcells, and epithelial cells all transmit PRRs on a surface level, in endosomes or in the cytoplasm of host cells. Sanctioning of these PRRs by PAMPs prompts setting off of intracellular hailing pathways, as MAPK (mitogen-started protein kinase) and NF-𝜅B (nuclear factor kappa-light-chain-enhancer of incited B cells) pathways, and finally to further developed record of countless characteristics drew in with have safe protections, including chemokines, cytokines, provocative center individuals, and antimicrobial peptides. Appropriately, PRRs are fundamental center individuals among intrinsic and adaptable safe responses.

Advertisement

3. Receptor molecules in Candida recognition

While comparing the human genome with murine genome; human genome encodes for ten TLR characteristics (TLR1–10) and murine genome encodes 12 i.e. TLR1 to TLR9 and TLR11 to TLR13. Each TLRs depicted as transmembrane type-1receptors having an enriched lucine extracellularly intermittent region which sees target PAMP and a Toll/interleukin-1 receptor-(TIR-) space containing cytoplasmic region that imparts the institution stimuli, which having closeness to the sort 1 interleukin-1 (IL-1) receptor. TLR family is a developmentally monitored gathering of PRRs that react to an assortment of bacterial, viral, and contagious PAMPs just as some endogenous components delivered when have cells are harmed. The extracellular areas of TLRs perceive an assortment of microbial PAMPs, including lipopolysaccharide (LPS), peptidoglycan, proteins (counting triacylated proteins and flagellin), and changed nucleic acids [53, 54, 55, 56, 57, 58].

3.1 Toll like receptors

3.1.1 TLR recognition of Candida

Key part for TLRs in host protection against fungal infection was initially identified when Drosophila inadequate in Toll receptor were seen to profoundly helpless to A. fumigatus disease [59]. Therefore by far most of the underlying antifungal insusceptibility research focused on how contagious cells were perceived. This provoked the distinctive verification of a couple of PRRs related with affirmation of different cell divider polysaccharides of parasites and C. albicans explicitly, including TLR2 (phospholipomannan), TLR4 (𝑂-associated mannan), and mannose receptor (MR) (𝑁-associated mannan) [46, 48, 60].

At last, these investigations finished in the disclosure of another PRR, dectin-1 (dendritic cell associated C-type lectin-1), who perceives parasitic 𝛽-1,3 glucan [61]. Outstandingly, these parasitic PRRs can work both freely and related to each other. For instance, dectin-1 and TLR2 act additionally to perceive contagious yeasts, with dectin-1 prompting phagocytosis while TLR2 initiates cytokine creation [62, 63, 64]. Dectin-1 likewise synergises with TLR4 flagging [64]. Moreover, TLR1 and TLR6 structure heterodimers with TLR2 [65] however do not seem to assume a significant part in C. albicans acknowledgment in a mouse model of intrusive candidiasis [66]. Obviously depending upon the coreceptor included, coligation of TLR2 may either update TLR2-subordinate responses [67] or change its PAMPs distinction concerning the circumstance with galectin-3 [68].

Even so these are standard receptors utilized by macrophages and neutrophils to see C. albicans, various receptors have moreover been perceived inclusive of dectin-2 [69], mincle (macrophage inducible CTL) [70], Dendritic cell specific intercellular grasp particle 3- getting nonintegrin (DC-SIGN ) [71, 72], and galectin-3 [68]. The piece of these PRRs is correct now not totally settled; regardless, dectin-2 and DC-SIGN are perceived to assume a significant part in the acknowledgment of high mannose structures [73] and galectin-3 in the acknowledgment of 𝛽-1,2 mannosides [68].

Curiously, galectin-3 coimmunoprecipitates accompanied by dectin-1 [74], which recommends that galectin-3 can work with associations among TLR2 and dectin-1 flagging. TLR acknowledgment of other medicinally significant growths have likewise been concentrated yet are less very much described, despite the fact that apparently TLR3 perceives A. fumigatus conidia and TLR4 perceives Cryptococcus neoformans glucuronoxylomannan, with TLR9 perceiving A. fumigatus, C. albicans and C. neoformans [75].

3.1.2 TLR signaling

PAMP acknowledgment of TLRs brings about enactment of flagging cascade intracellularly (Figure 2) through connection of the cytoplasmic TIR spaces with various connector proteins: myeloid separation essential reaction quality (88) (MyD88), MyD88-connector like (MAL), TIR-area containing connector initiating interferon-𝛽 (TRIF), and TRIF-related connector atom (TRAM) [53, 54, 55, 56, 57, 58, 76, 77, 78, 79]. This TLR-adapter interaction ends up in the activation of the IRAK ( IL-I receptor associated kinase) proteins and TRAF6 (TNF receptor associated factor-6). As a result it ends up in activation of the main signaling pathways together with NF-𝜅B, MAPK, and IRF (interferon regulative factor) pathways. MAPK activation contains 3 alleyways: p38, JNK (c-Jun N-terminal kinase), and ERKI/2 (extracellular signal-regulated kinaseI/2). Finally, signaling pathway induction ends up in the activation and nuclear localisation of transcription factors as well as NF-𝜅B, AP-I (activating macromolecule I), and IRF-3 and IRF-7. the result of this activation cascade is to induce organic phenomenon and secretion of varied proteins concerned in immune defense as well as cytokines, chemokines, antimicrobial peptides, and alternative inflammatory mediators, all of that operate to stimulate innate and reconciling responses of immune system. It thought to be noted that the overwhelming majority of studies shaping the TLR-mediated pathways are performed victimization myeloid or humor cells, however elaborated analysis of TLR- mediated pathways in alternative cell varieties, and specifically animal tissue cells, could nonetheless establish novel and strange mechanisms of infectious agent (fungal) recognition and management at membrane surfaces.

Figure 2.

Signal pathway activation by TLRs and CLRs. TLRs and CLRs activate MAPK and NF-𝜅B signal pathways to varying extents, thereby allowing different innate immune responses to be generated. TLRs utilize TIR-domain containing adapter proteins such as MyD88, mal, TRAM, and TRIF. CLRs signal using ITAM domains within their cytoplasmic region (e.g., dectin-1) or associate with an ITAM-containing transducing protein (e.g., dectin-2 with FcR𝛾). Dectin-1 utilizes Src kinases and Syk kinase to activate a complex containing CARD9, MALT1, and Bcl1o to activate the downstream signal pathways. Figure adapted from [47].

3.1.3 Role of TLRs during Candida infection

Although animals missing the TLR signaling adaptor protein MyD88 are vulnerable to fungal infection [46, 80, 81, 82], the exact role of particular TLR receptors in fighting Candida infections is unclear. This is most likely because of contrasts in examination plan, where diverse contagious species, morphotypes, and courses of contamination have been surveyed [52]. Thusly, contemplates utilizing TLR knockout mice have uncovered critical contrasts in the putative jobs of various TLRs in fundamental or mucosal insusceptible reactions against contagious contaminations [83]. For instance, while a few examinations demonstrate that TLR2 and TLR4 impact vulnerability to murine scattered candidiasis [82, 84, 85, 86], not all investigations support this attestation [87, 88]. TLR7 might be needed for parasitic RNA acknowledgment in the autophagosome, which is needed for IFN-𝛽 discharge and is related with delayed C. glabrata contamination [89]. TLR9 perceives C. albicans DNA (unmethylated CpG arrangements) bringing about cytokine creation in dendritic cells [90]; notwithstanding, TLR9 knockout mice do not seem, by all accounts, to be more helpless to C. albicans contamination, notwithstanding delivering diminished degrees of IL-I2 and expanded measures of IL-4 and IL-I0 [82, 90, 91, 92]. Outstandingly, explicit TLRs (TLR2, TLR4, TLR6, and TLR9) seem to hold various jobs relying upon which arm of the inborn invulnerable reaction they draw in with, for instance, advancement of versatile reactions by working with antigen show in dendritic cells [93].

A few examinations have related normal hereditary variations (polymorphisms) in TLR qualities with vulnerability or inclination to foundational candidiasis or constant mucocutaneous candidiasis (CMC). These recollect polymorphisms for TLRI (R80T , N248S , and S602I) [94, 95] and TLR3 (L4I2F) [96, 97]. Polymorphisms in TLR4 (D299G) and TLR2 (D753Q) have moreover been perceived as possible frailty markers for basic candidiasis [98] yet these could not be approved in a greater report [95]. As of now, a large portion of the information accessible recommends a solid part for TLRs in antifungal protection however recognizing explicit jobs for each TLR has been over shadowed by repetitive signs instigated by other PRRs [94].

3.2 C-type lectin receptors

CLRs (C-type lectin receptors) are a diverse restriction protein family defined by the presence of an extracellular carb acknowledgment space (CRD) or a C-type lectin like area (CTLD) [99]. The job of CLRs in antifungal insusceptibility has been the subject of serious investigation as of late and a few key CLRs have now been shown to show basic capacities in Candida acknowledgment, take-up, and executing and furthermore add to the commencement and additionally tweak of the resistant reaction to organisms [46, 100, 101]. By and by, the key CTLs in Candida affirmation appear, apparently, to be dectin-I, dectin-2, and MR. CLRs signal through incitation of ITAM/ITIM (immunoreceptor tyrosine-based actuation/restraint theme) cytoplasmic areas (Figure 3). This can be done by using their own cytoplasmic area, as dectin-I does, or by using coreceptor cytoplasmic spaces, as DAPI2 (DNAX actuation protein of I2 kDa) and FcR (Fc receptor gamma chain) do, as dectin-2 does. The activation of numerous connections to those activated by TLRs, most notably Src family kinases including Src, Lyn, and Fyn, is triggered when CLRs are ligated. If we talk about dectin-I , it prompts initiation of spleen tyrosine kinase (SYK) and the downstream actuation of the CARD9/BclI0/MALTI (caspase enlistment space family/B cell CLL-lymphoma I0/mucosa related lymphoid tissue lymphoma movement quality I) flagging complex. Independent of the CLR pathways and connectors utilized, a definitive outcome is the enactment of comparative flagging pathways as those initiated by TLRs, overwhelmingly NF-𝜅B and MAPK, that are discussed below point.

Figure 3.

Signaling and damage pathways activated by C. albicanshyphae. C. albicanshyphal cells, when in sufficient quantities, are recognized by an unknown PRR mechanism that results in the activation of NF-𝜅B, MAPK, and PI3K pathways. MAPK signaling via p38 and ERK1/2 appears to discriminate between yeast and hyphal cells. Activation of p38 by hyphae leads to activation of the c-Fos transcription factor, which, in conjunction with the p65/p50 NF-𝜅B heterodimers and PI3K/AKT results in upregulation of cytokine and inflammatory mediator expression. Concurrently, activation of ERK1/2 signaling, results in stabilization of the MKP1 phosphatase, which deactivates p38 and JNK, hence acting as part of a negative feedback loop and preventing a potentially deleterious overreaction of theimmune system. Damage induced by hyphae appears to be mediated via JNK activation and prevented via the PI3K/AKT/mTor pathway.

3.2.1 Dectin-I

Dectin-I, (also called CLEC7a) is that the main CLR known as taking part in a serious role in fungous recognition by the host system [102] and may be a sort II transmembrane macromolecule that belongs to a subgroup of CLRs referred to as natural killer (NK) receptor-like CLRs. The target ligands of dectin-I are 𝛽-I,3 glucan polymers, that comprise a serious part (∼60%) of fungous cell walls. The intracellular region of dectin-I contains a changed ITAM motif containing one amino acid residue rather than the standard 2 (hence the terms hem-ITAM or hemi- ITAM). Activation of the dectin-I results in phosphorylation of this domain and phosphorylation of SYK and activation of the BclI0- CARD9-MALTI complicated as mentioned on top of. This results in activation of each the canonical and noncanonical NF-𝜅B pathways [103] further as nuclear issue of activated T cells (NFAT) pathway [104]. Dectin-I can even induce signaling via Raf-I in an exceedingly SYK -dependent fashion [103] and is related to phospholipase C and A2 activation [50]. one in all the most important functions of dectin-I binding seems to be the induction of bodily process [105]. However, a singular feature of dectin-I is its ability to be activated or suppressed by its target matter. to completely activate dectin-I, cells got to be exposed to insoluble 𝛽-glucan particles. Notably, exposure of dectin-I to soluble 𝛽-glucan seems to dam activation. This appears to ensue to the apparent form type a vegetative cell conjunction,“whereby phosphatases that usually suppress ITAM motifs are accumulated. This exclusion later permits the phosphorylation of the intracellular hem-ITAM motif [106], thereby sanctioning bodily process. Dectin-I has additionally been shown to synergise with each TLR2 and TLR4, leading to the induction of tumor necrosis factor (TNF), IL-I0, transforming growth factor (TGF) and dendritic cell maturation [107, 108, 109]. In view of the fact that the 𝛽 - I,3 glucan polymers that are the main components of the fungal cell wall, and a strong activation of the immune system, dectin-I plays an important role in inducing antifungal activity of the host. This may also explain why some of the mold surface structure of “the mask” -I.3 glucan from the immune system. For example, Histoplasma capsulatum, masks are 𝛽-I,3 glucan, with a low - 𝛼-I,3 glucan [110] and it seems likely that the C. albicans hyphae of 𝛽-I,3 glucan has been covered over by layers of 𝑁 - 𝑂 - linked mannoproteins in order to prevent the discovery of the dectin-I. However, the yeast is in the form of C. albicans, while 𝑁 - 𝑂 - linked mannoproteins present in the underlying 𝛽 -glucan layer is exposed in the developing gut, which dectin-I in order to be recognized. Thus, it could be concluded that the most important role of dectin-I in the control of the yeast form of candidiasis (thrush). In addition, 𝛽 - glucan, which has been in the hyphal cell wall of C. albicans, it seems to be structurally different from the yeast 𝛽 - glucan [111] and, therefore, may not be immune to or understood by the dectin-I.

Although some studies have shown that the expression of dectin-I in the epithelial cells of the gastro- intestinal tract [112], and lung [113, 114], in oral epithelial cells express dectin-1 [115, 116]. What’s interesting is that dectin-I expression appears to be reduced in the presence of live C. albicans cells [116], and it is not affected by the dectin-I ligands [115, 117]. This suggests that dectin-I is likely to play only a minor role in the detection of C. albicans epithelial cells. Studies carried out with the help of dectin-I knockout mice have provided mixed data sets for the C. albicans systemic infection models, to demonstrate these differences, [118] and increased mortality [119] depending on the study, the C. albicans strain used. On the one hand, it is the work for the dectin-I is supported by the consideration that CARD9 knockout mice are susceptible to the most important infection [120] and in patients with head-and-CARD9 immunity and are particularly vulnerable to both the lining and the main foundation candidiasis [121]. In addition, another study, it has been the study of the normal function of the genetic polymorphism in CARD9 (SI2N), to CADR9, especially candidiasis, it is recommended that the method of fixing of the 𝛽 - glucan may be excessive for the first invulnerability of C. albicans [122]. However, recent studies have shown the potential role of dectin-I in the maintenance of tissue health. Dectin-I−/− the mice showed greater severity of the disease, at least one more commonly, however, this weight can be reduced by the removal of fungal and bacterial flora in [123]. Histologically, extensive infestations of fungi have been recorded from the underlying tissue, which was not seen in wild-type mice. Clinical trial data have shown that a subgroup of patients with ulcerative colitis, especially in aggressive disease, and shows a common single-nucleotide polymorphisms (rs2078I78 in dectin-I, possibly indicating a requirement for functional dectin-I receptors, and to maintain, mucosal health, in a commensal state [123]. However, the role of dectin-I in the intramucosal infections, it is far from clear, as recent studies in mice have demonstrated that dectin-I does not play an important role in the control of gastro-intestinal colonization by C. albicans [124]. In particular, it is well known that, in humans, mutations in the stop codon (Tyr238X in dectin-I is associated with an increased risk of developing mucocutaneous fungal infections, with an increased colonization of the oral cavity and the gastrointestinal (gi) tract and vulvovaginal candidiasis (thrush) infection (RVVC) [125, 126]. In another case, we obtained that the dectin-I polymorphism (I223S) was associated with oropharyngeal candidiasis (OPC) the susceptibility of West Africa, a group of HIV-positive patients [127]. That is why, even though the great one, the precise role of dectin-I in the susceptibility to candida infection is still unclear and requires further investigation.

3.2.2 Dectin-2

Dectin-2 (otherwise called CLEC6a) is a sort II transmembrane protein however is enacted contrastingly to dectin-I. Dectin-2 comes up short on an intracellular flagging area [128] and requirements to dimerise with FcR𝛾, which has an intracellular flagging space, to send a sign [69]. In myeloid cells and fiery monocytes, dectin-2 perceives high mannose structures that are normal to numerous parasites and ties to hyphae with higher proclivity than to yeast [129, 130]. This may clarify why dectin-2 inadequate mice are helpless to C. albicans contamination be that as it may, strangely, not C. neoformans [130, 131]. Dectin-2 may moreover recognize 𝛼-mannosyl linkages [132]. Dectin-2 may activate a number of cytokines and chemokines via NF-B, MAPK, SYK, CARD9-BclI0-MaltI, and PKC, as well as initiate the NLRP3 (NOD-like receptor family, pyrin region containing 3) inflammasome and a respiratory burst [69, 133]. Furthermore, dectin-2 may have a role in protecting against C. glabrata illnesses, since dectin-2/lacking mice were more susceptible to C. glabrata infections, indicating a poor transmittable choice in kidneys [134].

3.2.3 Dectin-3

Dectin-3 (additionally called CLECsf8, MCL, or CLEC4d) was as of late distinguished and seems to shape heterodimers with dectin-2 to perceive 𝛼-mannans on the outside of C. albicans hyphae, prompting NF-𝜅B enactment [135]. Strikingly, dectin-3−/− mice were exceptionally helpless to C. albicans disease. Contrasted and their particular homodimers, dectin-2/3 heterodimers bound 𝛼-mannans all the more viably, prompting strong incendiary reactions. This recommends that distinctive CLRs may shape an assortment of hetero and homodimers that may give diverse affectability and variety to have cells to identify different contagious contaminations.

3.2.4 DC-SIGN

DC-SIGN (otherwise called CD209) is another sort II transmembrane receptor that is communicated dominatingly on dendritic cells and macrophages. Dendritic Cell-Specific Intercellular adhesion molecule-3-Grabbing Non-integrin) also known as CD209 (Cluster of Differentiation 209) is a protein which in humans is encoded by the CD209 gene. DC-SIGN is a C-type lectin receptor present on the surface of both macrophages and dendritic cells Nonetheless, the part of DC-SIGN in antifungal invulnerability is muddled [101], in spite of the fact that DC-SIGN seems to perceive high (𝑁-connected) mannose containing glycoproteins and actuate IL-6 creation [71, 136]. Albeit the part of DC-SIGN in the endocytosis and take-up of microbes to advance antigen show is all around recorded [136, 137], its job in phagocytosis is sketchy [71, 136].

3.3 Mannose receptor

The MR (or called CD206) is a prototypical kind I transmembrane protein that is transcendently communicated on macrophage and dendritic cells. MR receptor ties a few starch particles, including extended 𝑁-connected mannans, N-acetylglucosamine, glucose, and fucose [138]. Thus, MR can perceive numerous contagious, bacterial, and viral pathogens. MR needs regular intracellular flagging spaces despite the fact that ligation actually prompts an assortment of cell reactions, including signal pathway acceptance, phagocytosis, advancement of antigen show to T cells, and cytokine discharge [63, 136, 137, 138, 139, 140]. For instance, the MR is enlisted to the phagosome after C. albicans ingestion and actuates intracellular flagging and cytokine creation [141]. MR may likewise be needed for the enlistment of defensive ThI7 reactions in C. albicans contamination [140] however may repress cytokine creation because of different organisms, for instance, Pneumocystis carinii [142]. Remarkably, MR inadequacy does not seem to present helplessness to C. albicans foundational disease [143] as it does to C. neoformans [144], albeit minor changes in parasitic weights can be noticed [143]. In oral epithelial cells, MR impeding does not modify the discharge of IL-6, IL-8, and GM-CSF upon incitement with Candida cell divider parts [117]. As of now, there is no conclusive part for MR in mucosal antifungal host safeguards.

3.4 Nod-like receptors (NLRs) and inflammasomes

NLRs are a group of intracellular PRRs portrayed by leucine rich rehashes and a nucleotide-restricting area that identify PAMPs present in the cell cytoplasm. Like TLRs and CTLs, NLRs perceive microbial items yet they additionally perceive have determined threat signals or alarmins [145]. There are now 23 human NLRs and 34 mouse NLRs identified [146]. Inflammasomes are huge multimeric protein structures framed by NLRs and two distinct proteins, ASC (apoptosis-related spot like protein containing a CARD) and procaspase-I (procysteine-subordinate aspartate-coordinated protease I). The inflammasome’s main function is to convert procaspase-I to dynamic caspase-I, which causes young cells that are friendly to IL-I and supportive of IL- I8 to produce IL-I and IL-I8 [147]. Despite the fact that C. albicans is not recognized by NLRCI (NLR family CARD space containing protein I) or NLRC2 [148], it is known to activate inflammasomes fusing NLRP3 (NACHT , LRR, and PYD spaces containing protein 3) [149] and NLRC4 [150], resulting in the production of IL-I.

Surprisingly, NLRP3 is strongly expressed in nonkeratinizing epithelia, such as the oral cavity and throat [151], suggesting a possible role for NLRP3 in parasitic recognition in oral epithelial cells, which is supported by studies showing increased IL- I and IL- I8 levels in response to C. albicans stimulation [115, 152, 153, 154, 155]. Mice missing NLRP3 appear to be susceptible to candidiasis [156], but mice lacking IL-I receptor type I (IL- IRI), IL-I8, or caspase-I exhibit distinct contagious contamination helplessness profiles [157]. Strikingly, IL-I𝛽 (and IL-I𝛼) lacking mice show expanded mortality during scattered candidiasis [158]. Late reports have likewise recognized a significant part for NLRP3 along with TLR2 and dectin-I in forestalling dispersal of C. albicans in a murine model of oral contamination [159]. Steady with a part for NLRP3 inmucosal security [160], deficient NLRP3 actuation expands C. albicans colonization in the gut and fuels Crohn’s illness [161], and a length polymorphism in intron 4 of the quality (CIASI) that codes for NLRP3 inclines patients to RVVC [162]. Nevertheless, the full degree of the practical jobs for NLRs and inflammasomes in antifungal host safeguards is as yet not completely comprehended.

Advertisement

4. Protein involves in pathogenesis

4.1 Mincle

Mincle (also known as CLEC4e or CLECsf9) is a type II transmembrane protein that transmits its signal after dimerizing with the FcR connector protein [128]. Macrophages, monocytes, neutrophils, myeloid dendritic cells, and certain B cell subsets all communicate mincle, while plasmacytoid dendritic cells, T cells, and NK cells do not [133]. Mincle binds -mannans-containing starch structures [143, 163] and detects C. albicans [70, 164, 165], Malassezia spp. [163], and Fonsecaea pedrosoi, the chromoblastomycosis causative pathogen [166]. As with dectin-2, mincle is not believed to be needed for phagocytosis [70] yet adds to the acceptance of cytokines and chemokines by means of NF-𝜅B, MAPK, SYK, CARD9-BclI0-MatIt, and PKC𝛿 [133, 163]. In spite of the fact that mincle-incited reactions have all the earmarks of being MyD88 autonomous, mincle may synergise with TLRs to instigate fiery cytokines and the respiratory burst [167].

4.2 Soluble proteins in Candida recognition

The supplement course assumes a significant part in have protection against parasitic microorganisms and is quickly enacted in light of host attack by Candida [168, 169, 170]. Candida actuates each of the three known pathways (old style, elective, and mannose-restricting lectin (MBL)) with nobody clear pathway overwhelming the reaction [171]. Given that the Candida cell surface is covered with a bounty of manno proteins, it is not astonishing that Candida microorganisms are viable at actuating the MBL pathway, which seems significant for opsonisation, phagocytosis, and other supplement capacities [172, 173]. The connection between enacted C3b and the supplement receptor CR3 is generally needed for the uptake of Candida cells by phagocytes [174]. C. albicans cell divider proteins (e.g., GpmI, PraI, and Gpd2) can possibly tie supplement segments, for example, Factor H, FHL-I, C4BP and plasminogen from human plasma that meddle with phagocytic opsonisation and take-up [168, 170, 175, 176, 177, 178, 179, 180]. For example, restricting of Pra1 to factor H and FHL-1 most likely includes an avoidance methodology including the hindrance of C3 cleavage into opsonic and anaphylatoxic parts, in this manner forestalling acknowledgment and take-up by phagocytes [181]. C5 is likewise significant in Candida diseases since mice that need practical C5 quality duplicates are vulnerable to obtrusive foundational contaminations [182, 183, 184, 185]. C5 insufficiency is related with expanded creation of proinflammatory cytokines (TNF𝛼 and IL-6) and fast parasitic replication in organs that can prompt cardiovascular disappointment [186, 187]. Sanctioning of C5 prompts the improvement of C5b, which consequently triggers the plan of the film attack complex (MAC). Despite the fact that affidavit of MAC on the outside of C. albicans does not bring about fungicidal movement, presumably because of the thickness of the parasitic cell divider, it might work with the incitement of phagocytes and ensuing arrival of terminal supplement segments from these phones. Curiously, as no impact on irritation is recognized in C3 insufficient mice, this may recommend a generally C3-free preparing of C5 in foundational C. albicans disease [188]. After phagocytosis, the oxidative burst is set off which prompts contagious executing, a cycle that can be hindered with monoclonal antibodies to forestall C3b-CR3 associations. C3b-CR3 contact also appears to be crucial for lymphocyte hyphal formation and cytokine production [189]. MBL has also been linked to the inhibition of Candida development [190] and the enhancement of TNF release from Candida-infected monocytes [191]. C3a, an anaphylatoxin released by C3 during supplement enactment, may have direct antifungal activity independent of its chemotactic effect [192]. These findings suggest that complement activation is critical in the host’s defense against C. albicans infections. The reader is directed to the following reviews [168, 170] for further in-depth information on the involvement of complement in Candida infections.

Advertisement

5. Cellular responses to Candida

5.1 Neutrophils

Neutrophils are a key effector cell in intrinsic insusceptibility, and they play a dual role in antifungal responses. First, they phagocytose and destroy contaminated Candida cells (below), and then, via cross communication with epithelial cells, they indirectly assist in mucosal protection (tended to above). TLRs and CTLs help neutrophils phagocytose nonopsonized Candida, while CR3 and the Fc receptor (FcR) help them phagocytose opsonized Candida [193]. Once phagocytosed, Candida is killed both inside and outside the cell through oxidative and nitrosative mechanisms, but fungicidal movement varies across Candida species [194, 195]. Preformed cytoplasmic granules interweave with the phagosome intracellularly, although unlike macrophages, no substantial pH changes occur [196]. Antimicrobial proteins found in neutrophil granules include defensins, lactoferrin, lysozyme, myeloperoxidase, and elastase [197], all of which can be transported into the extracellular environment. Candida’s phagocytic execution requires oxidative processes. During the oxidative burst, neutrophils create reactive oxygen species (ROS), which needs the NADPH oxidase catalyst complex to assemble in the cytoplasmic and phagosomal film [198]. First, the superoxide extremist is formed, which is subsequently dismutated to hydrogen peroxide, an oxidative and harmful particle [199].

Then, myeloperoxidase uses hydrogen peroxide to create hypochlorous acid, which is moreover an exceptionally oxidative particle that responds with natural amines to frame chloramines that have further antimicrobial stuffs [193, 200]. Candida’s phagocytic execution is further aided by reactive nitrogen species (RNS) [193]. When neutrophils are activated, they produce nitric oxide (NO) from arginine and oxygen via an enzyme called inducible nitric oxide synthase (iNOS). NO is extremely sensitive, and it is converted to peroxynitrite, which is then reduced to nitrogen dioxide and a hydroxyl radical. Because iNOS is restricted to the intracellular compartment, RNS production is restricted to the intracellular compartment [199]. The creation of neutrophil extracellular catches (NETs) [201, 202], which are formed during a unique sequence of neutrophil cell death known as NETosis, is another more recently found way of Candida executing. Similar to serine proteases, antimicrobial peptides (e.g., calprotectin), and other microbicidal chemicals, the neutrophil “explodes,” unleashing a snare of chromatin fibrils coated with the neutrophil’s material. Candida spp. are well-versed in surviving the oxidative, nitrosative, osmotic, and restorative nerves encountered during interactions with neutrophils. Because of the weights, many cycles, features, and proteins are altered within the organism. These include upregulation of transporters (e.g., oligopeptide, ammonium, and iron), use of alternative carbon and nitrogen sources and metabolic cycles (e.g., glycolysis, glyoxylate, unsaturated fat, and amino destructive), and detoxification of neutrophil oxidative/nitrosative butchering instruments. (e.g., catalase, superoxide dismutases, and nitric oxide dioxygenase). In any event, these nuances are beyond the scope of this examination, and the reader is directed to a later examination that focuses on the Candida reaction to neutrophils [193, 203].

5.2 Macrophages

Macrophages can function as phagocytic cells as well as antigen-presenting cells capable of activating T lymphocytes. Upon activation, macrophages divide into two phenotypically and functionally distinct subsets, M1 and M2, based on the cytokine milieu in which they are initiated [204, 205, 206]. The M1 total is derived from receptiveness to the T colleague (Th)1 cytokine IFN, whereas the M2 total is derived from receptiveness to Th2 cytokines, IL-4 and IL- 13. M1 macrophages are microbic and proinflammatory, whereas M2 macrophages are involved in wound healing and extracellular network upgradation. Macrophages, like neutrophils, see and phagocytoze nonopsonised Candida via TLRs and CTLs, and opsonised Candida via CR3 and FcR [193, 207]. Nonetheless, macrophage phagosome formation differs from neutrophil phagosome development in that macrophage phagosomes follow the endocytic development route and grow into phagolysosomes with a distinctive acidic pH that promotes compound activity, such as cathepsin D [208]. M1 macrophages use both oxidative and nitrosative executing components (as seen above for neutrophils), but they also use the RNS, NO, to directly kill phagocytosed Candida via the translocation of iNOS. TNF and the chemokines CXCL9 and CXCL10 are also released by M1 macrophages [209]. These chemokines act as ligands for the CXCR3 receptor, which is found on Th1 cells and NK cells, attracting resistant cells to contamination sites.

M2 macrophages, then again, advance contagious ingenuity inside the macrophage, giving an instrument to invulnerable avoidance. M2 macrophages additionally express more significant levels of MR (CD206) bringing about expanded phagocytosis of Candida [210]. Correspondingly, the arginase-1 (Arg1) quality is additionally expanded in articulation, which rivals iNOS for a similar substrate (arginine), consequently diminishing NO levels [211]. This is additionally exacerbated by decreased degrees of TNF𝛼 creation in M2 macrophages. In light of this, macrophages anticipate playing an important role in Candida protection, but this is contingent on the Candida strain assisting the macrophage [212]. Candida spp., like neutrophils, are believed to rely on relative adaptations to survive in macrophages. C. albicans and C. glabrata have been shown to alter metabolic requirements by using alternative carbon sources, upregulating impetuses for gluconeogenesis, glyoxylate cycle, and -oxidation of unsaturated lipids, and downregulating protein synthesis and glycolysis [193, 207]. This combines the formation of catalase and superoxide dismutases for extracellular ROS detoxification [213] and the outflow of flavohemoglobin impetuses for intracellular RNS butchering [214]. Concerning C. albicans, intracellular dealing additionally seems unusual and the growth may repress both lysosomal fermentation and NO delivery [215]. For additional subtleties the peruser is guided to ongoing surveys that emphasis on the Candida reaction to macrophages [207].

Besides these receptors molecules, actively participated proteins and cellular mechanism system there is a lot of others factors in these mechanisms are linked like adhesins and invasins, biofilm formation, contact sensing and thigmotropism, secreted hydrolases, pH-sensing and its regulation, environment and metabolic adaptation, small HSPs, metal acquisition. So, for a complete understanding these factors also play significant role in pathogenicity mechanism of C. albicans.

Advertisement

6. Conclusion

This chapter has discussed the pathogenicity mechanism along with host and cellular responses in Candida species. Host reactions to Candida are profoundly assorted because of the assortment of contagious PAMPs and antigens perceived by various safe cells at different disease destinations. Many inquiries have been conducted on this important topic, particularly with C. albicans, and thus we have obtained a much improved understanding of the appropriate structures of the PAMPs & PRRs. Still, further analysis is needed in order to attain insight into the complex communication between PAMPs and the corresponding receptors. Definitely, co-stimulation via multiple PAMP–PRR interactions may increase together the sensitivity as well as the specificity of the immune recognition process.

References

  1. 1. Vincent JL, Rello J, Marshall J, Silva E, Anzueto A, Martin CD et al (2009) EPIC II Group of Investigators. International study of the prevalence and outcomes of infection in intensive care units. JAMA 302:2323-2329
  2. 2. Ingham CJ, Boonstra S, Levels S, de Lange M, Meis JF, Schneeberger PM (2012) Rapid susceptibility testing and microcolony analysis of Candida spp. cultured and imaged on porous aluminium oxide. PLoS ONE 7:e33818
  3. 3. Correia A, Sampaio P, Vilanova M, Pais C (2015) Candida albicans: clinical relevance, pathogenesis, and host immunity. In: Sing SK (ed) Human emerging and re-emerging infections: viral and parasitic infections, vol 1. John Wiley and Sons, New Jersey, pp 926-952
  4. 4. Limon JJ, Skalski JH, Underhill DM (2017) Commensal fungi in health and disease. Cell Host Microbes 22:156-165
  5. 5. De Rosa FG, Garazzino S, Pasero DC, Peri GD (2009) Invasive candidiasis and candidemia: new guidelines. Minerva Anaestesiologica 75:453-458
  6. 6. Negri M, Faria M, Guilhermetti E, Alves A, Paula C, Svidzinski T (2010) Hemolytic activity and production of germ tubes related to pathogenic potential of clinical isolates of Candida albicans. J Basic Appl Pharm. 31:89-93
  7. 7. Silva S, Negri M, Henriques M, Oliveira R, Williams DW, Azeredo J (2011) Adherence and biofilm formation of non- Candida albicans Candida species. Trends Microbio 19:241-247
  8. 8. Wisplinghoff H, Seifert H, Tallent SM, Bischoff T, Wenzel RP, Edmond MB (2003a) Nosocomial bloodstream infections in pediatric patients in United States hospitals: epidemiology, clinical features and susceptibilities. Pediatr Infect Dis J 22:686-691
  9. 9. Bongomin F, Gago S, Oladele R, Denning D (2017) Global and multi-national prevalence of fungal diseases-estimate precision. J Fungi 3:57
  10. 10. Dadar M, Tiwari R, Karthik K, Chakraborty S, Shahali Y, Dhama K (2018) Candida albicans-biology, molecular characterization, pathogenicity, and advances in diagnosis and control-an update. Microb Pathog 117:128-138
  11. 11. Caceres DH, Forsberg K, Welsh RM, Sexton DJ, Lockhart SR, Jackson BR et al (2019) Candida auris: a review of recommendations for detection and control in health care settings. J Fungi 5:111
  12. 12. Aslam B,Wang W, Arshad MI, Khurshid M,Muzammil S, Rasool MH et al (2018) Antibiotic resistance: a rundown of a global crisis. Infect Drug Resist 11:1645-1658
  13. 13. Kornitzer D (2019) Regulation of Candida albicans hyphal morphogenesis by endogenous signals. J Fungi 5:21
  14. 14. Wisplinghoff H, Elobers J, Geurtz L, Stefanik D, Major Y, Edmond MB et al (2014) Nosocomial bloodstream infections due to Candida spp. in the USA: species distribution, clinical features and antifungal susceptibilities. Int J Antimicrob Agents 43:78-81
  15. 15. Perez JC, Johnson AD (2013) Regulatory circuits that enable proliferation of the fungus Candida albicans in a mammalian host. PLoS Pathogen 9(12):e1003780
  16. 16. Jacobsen ID, Hube B (2017) Candida albicans morphology: still in focus. Expert Rev Anti Infect Ther 15:327-330
  17. 17. AokiW, Kitahara N,Miura N, Morisaka H, Yamamoto Y, Kuroda K et al (2011) Comprehensive characterization of secreted aspartic proteases encoded by a virulence gene family in Candida albicans. J BioChem 150:431-438
  18. 18. Seman BG, Moore JL, Scherer AK, Blair BA, Manandhar S, Jones JM et al (2018) Yeast and filaments have specialized, independent activities in a zebrafish model of Candida albicans infection. Infect Immun 86:e00415–e00418
  19. 19. Desai JV, Cheng S, Ying T, NguyenMH, Clancy CJ, Lanni F et al (2015) Coordination of Candida albicans invasion and infection functions by phosphoglycerol phosphatase Rhr2. Pathogens 4: 573-589
  20. 20. Kadosh D (2017) Morphogenesis in C. albicans. In: Prasad R (ed) Candida albicans: Cell Mol Biol. Springer, Cham
  21. 21. Han TL, Cannon RD, Villas-Boas SG (2011) The metabolic basis of Candida albicans morphogenesis and quorum sensing. Fungal Genet Biol 48:747-763
  22. 22. Monge RA, Román E, Nombela C, Pla J (2006) The MAP kinase signal transduction network in Candida albicans. Microbiology 152:905-912
  23. 23. Gong Y, Li T, Yu C, Sun S (2017) Candida albicans heat shock proteins and Hsps-associated signaling pathways as potential antifungal targets. Front Cell Infect Microbiol 7:520
  24. 24. Smith DA, Nicholls S,Morgan BA, Brown AJP, Quinn JA (2004) Conserved stress-activated protein kinase regulates a core stress response in the human pathogen Candida albicans.Mol Biol Cell 15:4179-4190
  25. 25. Hogan D, Sundrom P (2009) The Ras/Camp/PKA signaling pathways and virulence in Candida albicans. Future Microbiol 4: 1263-1270
  26. 26. Lin C-J, Wu C-Y, Yu S-J, Chen Y-L (2018) Protein kinase A governs growth and virulence in Candida tropicalis. Virulence 9(1):331-347
  27. 27. Inglis DO, Sherlock G (2013) Ras signaling gets fine-tuned: regulation of multiple pathogenic traits of Candida albicans. Eukaryot Cell 12:1316-1325
  28. 28. Lin CJ, Chen YL (2018) Conserved and divergent functions of the cAMP/PKA signaling pathway in Candida albicans and Candida tropicalis. J Fungi 4:68
  29. 29. DavisDA (2009) Howhuman pathogenic fungi sense and adapt to pH: the link to virulence. Curr Opin Microbiol 12:365-370
  30. 30. Brown A, Haynes K, Gow N, Quinn J (2012) Stress responses in Candida, 2nd edn. ASM Press, Washington, D.C., pp 225-242
  31. 31. Zhou Y, LiaoM, Zhu C, Hu Y, Tong T, Peng X et al (2018) ERG3 and ERG11 genes are critical for the pathogenesis of Candida albicans during the oral mucosal infection. Int J Oral Sci 10:9
  32. 32. de Oliveira SGC, Vasconcelos CC, Lopes AJO, de Sousa Cartagenes MDS, Filho AKDB, do Nascimento FRF et al (2018) Candida infections and therapeutic strategies: mechanisms of action for traditional and alternative agents. Front Microbiol 9:1351
  33. 33. Dantas SA, Lee KK, Raziunaite I, Schaefer K, Wagener J, Yadav B et al (2016) Cell biology of Candida albicans-host interactions. Curr Opin Microbiol 34:111-118
  34. 34. Schonherr FA, Sparber F, Kirchner FR, Guiducci E, Trautweinweidner K, Gladiator A et al (2017) The interspecies diversity of C. albicans triggers qualitatively and temporally distinct host responses that determine the balance between commensalism and pathogenicity. Mucosal Immunol 10:1335-1350
  35. 35. Braunsdorf C, LeibundGut-Landmann S (2018) Modulation of the fungal-host interaction by the intra-species diversity of C. albicans. Pathogens 7:11
  36. 36. Reedy JL, Filler SG, Heitman J (2010) Elucidating the Candida albicans calcineurin signaling cascade controlling stress response and virulence. Fungal Genet Biol 47:107
  37. 37. Liu S, Liu W (2015) Components of the canclium-calcinerium signaling pathways in fungal cells and their potential as antifungal targets. Eukaryot Cell 14:4
  38. 38. Yu Q, Jia C, Dong Y, Zhang B, Xiao C, Chen Y et al (2015) Candida albicans autophagy, no longer a bystander: its role in tolerance to ER stress-related antifungal drugs. Fungal Genet Biol 81:238-249
  39. 39. Shang-Jie Y, Ya-Lin C, Ying-Lie C (2015) Calcineurin signaling: lessons from Candida species. FEMS Microbiol 15:4
  40. 40. Wang L, Lin X (2012) Morphogenesis in fungal pathogenesis: shape, size and surface. PLoS Pathog 8:e1003027
  41. 41. Kim S, Nguyen QB,WolyniakMJ, Frechette G, Lehman CR, Fox BK et al (2018) Release of transcriptional repression through the HCR promoter region confers uniform expression of HWP1 on surfaces of Candida albicans germ tubes. PLoS ONE 13: e0192260
  42. 42. Sharma J, Rosiana S, Razzaq I, Shapiro RS (2019) Linking cellular morphogenesis with antifungal treatment and susceptibility in Candida pathogens. J Fungi 5:17
  43. 43. Sun JN, Solis NV, Phan QT, Bajwa JS, Kashlera H, Thompson A et al (2010) Host cell invasion and virulence mediated by Candida albicans Ssai. PLos Pathog 6:e1001181
  44. 44. Wächtler B,Wilson D, Haedicke K, Dalle F, Hube B (2011) From attachment to damage: defined genes of Candida albicans mediate adhesion, invasion and damage during interaction with oral epithelial cells. PLoS One 6:e17046
  45. 45. C. A. Janeway Jr. and R. Medzhitov, “Innate immune recognition,” Annual Review of Immunology, vol. 20, pp. 197-216, 2002
  46. 46. M. G. Netea, G. D. Brown, B. J. Kullberg, and N. A. R. Gow, “An integrated model of the recognition of Candida albicans by the innate immune system,“Nature ReviewsMicrobiology, vol. 6, no. 1, pp. 67-78, 2008
  47. 47. J. R. Naglik and D. Moyes, “Epithelial cell innate response to Candida albicans.,” Advances in dental research, vol. 23, no. 1, pp. 50-55, 2011
  48. 48. A. Roeder, C. J. Kirschning, R. A. Rupec, M. Schaller,G.Weindl, and H. C. Korting, “Toll-like receptors as key mediators in innate antifungal immunity,” Medical Mycology, vol. 42, no. 6, pp. 485-498, 2004
  49. 49. G. Weindl, J. Wagener, and M. Schaller, “Epithelial cells and innate antifungal defense,” Journal of Dental Research, vol. 89, no. 7, pp. 666-675, 2010
  50. 50. A. Plato, J. A. Willment, and G. D. Brown, “C-Type lectinlike receptors of the dectin-1 cluster: Ligands and signalling pathways,” International Reviews of Immunology, vol. 32, no. 2, pp. 134-156, 2013
  51. 51. D. J. Philpott, M. T. Sorbara, S. J. Robertson, K. Croitoru, and S. E. Girardin, “NOD proteins: regulators of inflammation in health and disease,” Nature Reviews Immunology, vol. 14, pp. 9-23, 2014
  52. 52. C. Bourgeois and K. Kuchler, “Fungal pathogens—a sweet and sour treat for toll-like receptors,” Frontiers in Cellular and Infection Microbiology, vol. 2, article 142, 2012
  53. 53. S. Akira, “Mammalian Toll-like receptors,” Current Opinion in Immunology, vol. 15, no. 1, pp. 5-11, 2003
  54. 54. K. Takeda, T. Kaisho, and S. Akira, “Toll-like receptors,” Annual Review of Immunology, vol. 21, pp. 335-376, 2003
  55. 55. S. Akira and K. Takeda, “Toll-like receptor signalling,” Nature Reviews Immunology, vol. 4, no. 7, pp. 499-511, 2004
  56. 56. K. Takeda and S. Akira, “TLR signaling pathways,” Seminars in Immunology, vol. 16, no. 1, pp. 3-9, 2004
  57. 57. T. Kawai and S. Akira, “Pathogen recognition with Toll-like receptors,” Current Opinion in Immunology, vol. 17, no. 4, pp. 338-344, 2005
  58. 58. T. Kawai and S. Akira, “TLR signaling,” Seminars in Immunology, vol. 19, no. 1, pp. 24-32, 2007
  59. 59. B. Lemaitre, E. Nicolas, L. Michaut, J. Reichhart, and J. A. Hoffmann, “The dorsoventral regulatory gene cassette spatzle/Toll/Cactus controls the potent antifungal response in Drosophila adults,” Cell, vol. 86, no. 6, pp. 973-983, 1996
  60. 60. T. Jouault, S. Ibata-Ombetta, O. Takeuchi et al., “Candida albicans phospholipomannan is sensed through toll-like receptors,” Journal of Infectious Diseases, vol. 188, no. 1, pp. 165-172, 2003
  61. 61. G. D. Brown, P. R. Taylor, D.M. Reid et al., “Dectin-1 is a major 𝛽-glucan receptor on macrophages,” Journal of Experimental Medicine, vol. 196, no. 3, pp. 407-412, 2002
  62. 62. K. M. Dennehy, J. A. Willment, D. L. Williams, and G. D. Brown, “Reciprocal regulation of IL-23 and IL-12 following coactivation of dectin-1 and TLR signaling pathways,” European Journal of Immunology, vol. 39, no. 5, pp. 1379-1386, 2009
  63. 63. M. G. Netea, N. A. R. Gow, C. A. Munro et al., “Immune sensing of Candida albicans requires cooperative recognition of mannans and glucans by lectin and Toll-like receptors,” Journal of Clinical Investigation, vol. 116, no. 6, pp. 1642-1650, 2006
  64. 64. G. Ferwerda, F. Meyer-Wentrup, B. Kullberg, M. G. Netea, and G. J. Adema, “Dectin-1 synergizes with TLR2 and TLR4 for cytokine production in human primary monocytes and macrophages,” Cellular Microbiology, vol. 10, no. 10, pp. 2058– 2066, 2008
  65. 65. A.Ozinsky, D.M.Underhill, J.D. Fontenot et al., “The repertoire for pattern recognition of pathogens by the innate immune system is defined by cooperation between Toll-like receptors,” Proceedings of the National Academy of Sciences of the United States of America, vol. 97, no. 25, pp. 13766-13771, 2000
  66. 66. M. G. Netea, F. Van De Veerdonk, I. Verschueren, J.W. M. Van DerMeer, andB. J. Kullberg, “RoleofTLR1 andTLR6 in thehost defense against disseminated candidiasis,” FEMS Immunology and Medical Microbiology, vol. 52, no. 1, pp. 118-123, 2008
  67. 67. S. P. Smeekens, F. L. van de Veerdonk, J. W. M. van der Meer, B. J. Kullberg, L. A. B. Joosten, andM. G.Netea, “The Candida Th17 response is dependent on mannanand 𝛽-glucan-induced prostaglandin E2,” International Immunology, vol. 22, no. 11, pp. 889-895, 2010
  68. 68. T. Jouault, M. El Abed-El Behi, M. Mart’ınez-Esparza et al., “Specific recognition of Candida albicans by macrophages requires galectin-3 to discriminate Saccharomyces cerevisiae and needs association with TLR2 for signaling,” Journal of Immunology, vol. 177, no. 7, pp. 4679-4687, 2006
  69. 69. K. Sato, X. Yang, T. Yudate et al., “Dectin-2 is a pattern recognition receptor for fungi that couples with the Fc receptor 𝛾 chain to induce innate immune responses,” The Journal of Biological Chemistry, vol. 281, no. 50, pp. 38854-38866, 2006
  70. 70. C. A. Wells, J. A. Salvage-Jones, X. Li et al., “The macrophageinducible C-type lectin, mincle, is an essential component of the innate immune response to Candida albicans,” Journal of Immunology, vol. 180, no. 11, pp. 7404-7413, 2008
  71. 71. A. Cambi, K. Gijzen, I. J. M. de Vries et al., “The C-type lectin DC-SIGN (CD209) is an antigen-uptake receptor for Candida albicans on dendritic cells,” European Journal of Immunology, vol. 33, no. 2, pp. 532-538, 2003
  72. 72. P.R.Taylor,G.D. Brown, J.Herre,D. L.Williams, J. A.Willment, and S.Gordon, “TheRole of SIGNR1 and the 𝛽-GlucanReceptor (Dectin-1) in the Nonopsonic Recognition of Yeast by Specific Macrophages,” Journal of Immunology, vol. 172, no. 2, pp. 1157-1162, 2004
  73. 73. M. G. Netea and B. J. Kullberg, “Epithelial sensing of fungal invasion,” Cell Host andMicrobe, vol. 8, no. 3, pp. 219-220, 2010
  74. 74. A. Esteban, M.W. Popp, V. K. Vyas, K. Strijbis,H. L. Ploegh, and G. R. Fink, “Fungal recognition is mediated by the association of dectin-1 and galectin-3 in macrophages,” Proceedings of the National Academy of Sciences of the United States of America, vol. 108, no. 34, pp. 14270-14275, 2011
  75. 75. L. Romani, “Immunity to fungal infections,” Nature Reviews Immunology, vol. 11, no. 4, pp. 275-288, 2011
  76. 76. L. B. Ivashkiv, “A signal-switch hypothesis for cross-regulation of cytokine and TLR signalling pathways,” Nature Reviews Immunology, vol. 8, no. 10, pp. 816-822, 2008
  77. 77. S. V. Tsoni and G. D. Brown, “𝛽-Glucans and dectin-1,” Annals of the New York Academy of Sciences, vol. 1143, pp. 45-60, 2008
  78. 78. E. F. Kenny and L. A. J. O’Neill, “Signalling adaptors used by Toll-like receptors: an update,” Cytokine, vol. 43, no. 3, pp. 342-349, 2008
  79. 79. L. A. J. O’Neill, “The interleukin-1 receptor/Toll-like receptor superfamily: 10 Years of progress,” Immunological Reviews, vol. 226, no. 1, pp. 10-18, 2008
  80. 80. E. Villam’on, D. Gozalbo, P. Roig et al., “Myeloid differentiation factor 88 (MyD88) is required formurine resistance to Candida albicans and is critically involved in Candida-induced production of cytokines,” European Cytokine Network, vol. 15, no. 3, pp. 263-271, 2004
  81. 81. C. Bourgeois,O.Majer, I. E. Frohner, L. Tierney, and K. Kuchler, “Fungal attacks on mammalian hosts: pathogen elimination requires sensing and tasting,” Current Opinion in Microbiology, vol. 13, no. 4, pp. 401-408, 2010
  82. 82. S. Bellocchio, C. Montagnoli, S. Bozza et al., “The contribution of the Toll-like/IL-1 receptor superfamily to innate and adaptive immunity to fungal pathogens in vivo,” The Journal of Immunology, vol. 172, no. 5, pp. 3059-3069, 2004
  83. 83. M. L. Gil andD. Gozalbo, “Role of toll-like receptors insystemic Candida albicans infections,” Frontiers in Bioscience, vol. 14, no. 2, pp. 570-582, 2009
  84. 84. M. G. Netea, C. A. A. van der Graaf, A. G. Vonk, I.Verschueren, J. W. M. Van der Meet, and B. J. Kullberg, “The role of tolllike receptor (TLR) 2 and TLR4 in the host defense against disseminated candidiasis,” Journal of Infectious Diseases, vol. 185, no. 10, pp. 1483-1489, 2002
  85. 85. M.G.Netea, R. Sutmuller, C.Hermann et al., “Toll-like receptor 2 suppresses immunity against Candida albicans through induction of IL-10 and regulatory T cells,” Journal of Immunology, vol.172, no. 6, pp. 3712-3718, 2004
  86. 86. M. G. Netea, N. A. R. Gow, L. A. B. Joosten, I. Verschueren, J. W. M. Van Der Meer, and B. J. Kullberg, “Variable recognition of Candida albicans strains by TLR4 and lectin recognition receptors,” Medical Mycology, vol. 48, no. 7, pp. 897-903, 2010
  87. 87. E. Villam’on, D. Gozalbo, P. Roig et al., “Toll-like receptor 2 is dispensable for acquired host immune resistance to Candida albicans in a murine model of disseminated candidiasis,” Microbes and Infection, vol. 6, no. 6, pp. 542-548, 2004
  88. 88. C. Murciano, E. Villamon, D. Gozalbo, P. Roig, J. E. O’Connor, and M. L. Gil, “Toll-like receptor 4 defective mice carrying point or null mutations do not show increased susceptibility to Candida albicans in a model of hematogenously disseminated infection,” Medical Mycology, vol. 44, no. 2, pp. 149-157, 2006
  89. 89. C. Bourgeois, O. Majer, I. E. Frohner et al., “Conventional dendritic cells mount a type I IFN response against Candida spp. requiring novel phagosomal TLR7-mediated IFN-𝛽 signaling,” Journal of Immunology, vol. 186, no. 5, pp. 3104-3112, 2011
  90. 90. A. Miyazato, K. Nakamura, N. Yamamoto et al., “Toll-like receptor 9-dependent activation of myeloid dendritic cells by deoxynucleic acids from Candida albicans,” Infection and Immunity, vol. 77, no. 7, pp. 3056-3064, 2009
  91. 91. F. L. van de Veerdonk, M. G. Netea, T. J. Jansen et al., “Redundant role of TLR9 for anti-Candida host defense,” Immunobiology, vol. 213, no. 8, pp. 613-620, 2008
  92. 92. C. Biondo, G. Signorino, A. Costa et al., “Recognition of yeast nucleic acids triggers a host-protective type I interferon response,” European Journal of Immunology, vol. 41, no. 7, pp. 1969-1979, 2011
  93. 93. J. Magarian Blander and R. Medzhitov, “Toll-dependent selection of microbial antigens for presentation by dendritic cells,” Nature, vol. 440, no. 7085, pp. 808-812, 2006
  94. 94. T. S. Plantinga, M.D. Johnson,W. K. Scott et al., “Human genetic susceptibility to Candida infections,” MedicalMycology, vol. 50, no. 8, pp. 785-794, 2012
  95. 95. T. S. Plantinga,M.D. Johnson,W.K. Scott et al., “Toll-like receptor 1 polymorphisms increase susceptibility to candidemia,” Journal of Infectious Diseases, vol. 205, no. 6, pp. 934-943, 2012
  96. 96. A. Nahum, H. Dadi, A. Bates, and C. M. Roifman, “The L412F variant of Toll-like receptor 3 (TLR3) is associated with cutaneous candidiasis, increased susceptibility to cytomegalovirus, and autoimmunity,” Journal of Allergy and Clinical Immunology, vol. 127, no. 2, pp. 528-531, 2011
  97. 97. A. Nahum, H. Dadi, A. Bates, and C. M. Roifman, “The biological significance of TLR3 variant, L412F, in conferring susceptibility to cutaneous candidiasis, CMV and autoimmunity,” Autoimmunity Reviews, vol. 11, no. 5, pp. 341-347, 2012
  98. 98. C. A. A. Van der Graaf, M. G. Netea, S. A. Morr’e et al., “Tolllike receptor 4 Asp299Gly/Thr399Ile polymorphisms are a risk factor for Candida bloodstream infection,” European Cytokine Network, vol. 17, no. 1, pp. 29-34, 2006
  99. 99. A. N. Zelensky and J. E. Gready, “The C-type lectin-like domain superfamily,” FEBS Journal, vol. 272, no. 24, pp. 6179-6217, 2005
  100. 100. S. E. Hardison and G. D. Brown, “C-type lectin receptors orchestrate antifungal immunity,” Nature Immunology, vol. 13, no. 9, pp. 817-822, 2012
  101. 101. J. A. Willment and G. D. Brown, “C-type lectin receptors in antifungal immunity,” Trends in Microbiology, vol. 16, no. 1, pp. 27-32, 2008
  102. 102. G. D. Brown, “Dectin-1 : a signalling non-TLR patternrecognition receptor,” Nature Reviews Immunology, vol. 6, no. 1, pp. 33-43, 2006
  103. 103. S. I.Gringhuis, J. denDunnen,M. Litjens et al., “Dectin-1 directs T helper cell differentiation by controlling noncanonical NF-𝜅B activation through Raf-1 and Syk,” Nature Immunology, vol. 10, no. 2, pp. 203-213, 2009
  104. 104. D. M. Reid, N. A. Gow, and G. D. Brown, “Pattern recognition: recent insights fromDectin-1,” Current Opinion in Immunology, vol. 21, no. 1, pp. 30-37, 2009
  105. 105. J. Herre, J. A. Willment, S. Gordon, and G. D. Brown, “The role of dectin-1 in antifungal immunity,” Critical Reviews in Immunology, vol. 24, no. 3, pp. 193-203, 2004
  106. 106. H. S. Goodridge, C. N. Reyes, C. A. Becker et al., “Activation of the innate immune receptor Dectin-1 upon formation of a ‘Phagocytic synapse’,” Nature, vol. 472, no. 7344, pp. 471-475, 2011
  107. 107. G. D. Brown, J. Herre, D. L. Williams, J. A. Willment, A. S. J. Marshall, and S. Gordon, “Dectin-1 mediates the biological effects of 𝛽-glucans,” Journal of ExperimentalMedicine, vol. 197, no. 9, pp. 1119-1124, 2003
  108. 108. S. Dillon, S. Agrawal, K. Banerjee et al., “Yeast zymosan, a stimulus for TLR2 and dectin-1, induces regulatory antigenpresenting cells and immunological tolerance,” Journal of Clinical Investigation, vol. 116, no. 4, pp. 916-928, 2006
  109. 109. B. N. Gantner, R. M. Simmons, S. J. Canavera, S. Akira, and D. M. Underhill, “Collaborative induction of inflammatory responses by dectin-1 and toll-like receptor 2,” Journal of Experimental Medicine, vol. 197, no. 9, pp. 1107-1117, 2003
  110. 110. C. A. Rappleye, L. G. Eissenberg, and W. E. Goldman, “Histoplasma capsulatum𝛼-(1,3)-glucan blocks innateimmune recognition by the 𝛽-glucan receptor,” Proceedings of the National Academy of Sciences of the United States of America, vol. 104, no. 4, pp. 1366-1370, 2007
  111. 111. D. W. Lowman, R. R. Greene, D. W. Bearden et al., “Novel structural features in Candida albicans hyphal glucan provide a basis for differential innate immune recognition of hyphae versus yeast,” The Journal of Biological Chemistry, vol. 289, pp. 3432-3443, 2014
  112. 112. P. J. Rice, E. L. Adams, T. Ozment-Skelton et al., “Oral delivery and gastrointestinal absorption of soluble glucans stimulate increased resistance to infectious challenge,” Journal of Pharmacology and ExperimentalTherapeutics, vol. 314, no. 3, pp. 1079-1086, 2005
  113. 113. S. E. Evans, P. Y. Hahn, F. McCann, T. J. Kottom, Z. V. Pavlovi’c, and A. H. Limper, “Pneumocystis cell wall 𝛽-glucans stimulate alveolar epithelial cell chemokine generation through nuclear factor-𝜅𝛽-dependent mechanisms,” The American Journal of Respiratory Cell and Molecular Biology, vol. 32, no. 6, pp. 490-497, 2005
  114. 114. H. Lee, J. Yuk, D. Shin, and E. Jo, “Dectin-1 is inducible and plays an essential role for mycobacteria-induced innate immune responses in airway epithelial cells,” Journal of Clinical Immunology, vol. 29, no. 6, pp. 795-805, 2009
  115. 115. D. L.Moyes, M. Runglall, C.Murciano et al., “A biphasic innate immune MAPK response discriminates between the yeast and hyphal forms of Candida albicans in epithelial cells,” Cell Host and Microbe, vol. 8, no. 3, pp. 225-235, 2010
  116. 116. D. L. Moyes, C. Shen, C. Murciano et al., “Protection against epithelial damage during Candida albicans infection is mediated by PI3K/Akt and mammalian target of rapamycin signaling,” Journal of InfectiousDiseases, vol. 209, no. 11,pp. 1816-1826, 2014
  117. 117. J. Wagener, G. Weindl, P. W. J. de Groot et al., “Glycosylation of Candida albicans cell wall proteins is critical for induction of innateimmune responses and apoptosis of epithelial cells,” PLoS ONE, vol. 7, no. 11,Article ID e50518, 2012
  118. 118. S. Saijo, N. Fujikado, T. Furuta et al., “Dectin-1 is required for host defense against Pneumocystis carinii but not against Candida albicans,” Nature Immunology, vol. 8, no. 1, pp. 39-46, 2007
  119. 119. P. R. Taylor, S. V. Tsoni, J. A. Willment et al., “Dectin-1 is required for 𝛽-glucan recognition and control of fungal infection,” Nature Immunology, vol. 8, no. 1, pp. 31-38, 2007
  120. 120. O.Gross, A.Gewies, K. Finger et al., “Card9 controls a non-TLR signalling pathway for innate anti-fungal immunity,” Nature, vol. 442, no. 7103, pp. 651-656, 2006
  121. 121. A. Puel, S. Cypowyj, J. Bustamante et al., “Chronic mucocutaneous candidiasis in humans with inborn errors of interleukin-17 immunity,” Science, vol. 332, no. 6025, pp. 65-68, 2011
  122. 122. D. C. Rosentul, T. S. Plantinga, M. Oosting et al., “Genetic variation in the dectin-1/CARD9 recognition pathway and susceptibility to candidemia,” Journal of Infectious Diseases, vol. 204, no. 7, pp. 1138-1145, 2011
  123. 123. I. D. Iliev, V. A. Funari, K. D. Taylor et al., “Interactions between commensal fungi and the C-type lectin receptor dectin-1 influence colitis,” Science, vol. 336, no. 6086, pp. 1314-1317, 2012
  124. 124. S. Vautier, R. A. Drummond, P. Redelinghuys, G. I. Murray, D. M. MacCallum, and G. D. Brown, “Dectin-1 is not required for controlling Candida albicans colonization of the gastrointestinal tract,” Infection and Immunity, vol. 80,no. 12, pp. 4216-4222, 2012
  125. 125. B. Ferwerda, G. Ferwerda, T. S. Plantinga et al., “Human dectin-1 deficiency and mucocutaneous fungal infections,” The New England Journal ofMedicine, vol. 361, no. 18, pp. 1760-1767, 2009
  126. 126. T. S. Plantinga, W. J. F. M. Van Der Velden, B. Ferwerda et al., “Early stop polymorphism in human DECTIN-1 is associated with increased candida colonization in hematopoietic stem cell transplant recipients,” Clinical Infectious Diseases, vol. 49, no. 5, pp. 724-732, 2009
  127. 127. T. S. Plantinga, O. J. M. Hamza, J. A. Willment et al., “Genetic variation of innate immune genes in HIV-infected African patients with or without oropharyngeal candidiasis,” Journal of Acquired ImmuneDeficiency Syndromes, vol. 55,no. 1, pp. 87-94, 2010
  128. 128. L.M. Grahamand G. D. Brown, “TheDectin-2 family of C-type lectins in immunity and homeostasis,” Cytokine, vol. 48, no. 1-2, pp. 148-155, 2009
  129. 129. E. P. McGreal, M. Rosas, G. D. Brown et al., “The carbohydrate recognition domain of Dectin-2 is a C-type lectin with specificity for high mannose,” Glycobiology, vol. 16, no. 5, pp. 422-430, 2006
  130. 130. S. Saijo, S. Ikeda, K. Yamabe et al., “Dectin-2 recognition of 𝛼-mannans and induction of Th17 cell differentiation is essential for host defense against Candida albicans,” Immunity, vol. 32, no. 5, pp. 681-691, 2010
  131. 131. M. J. Robinson, F. Osorio, M. Rosas et al., “Dectin-2 is a Syk coupled pattern recognition receptor crucial forTh17 responses to fungal infection,” Journal of Experimental Medicine, vol. 206, no. 9, pp. 2037-2051, 2009
  132. 132. N. Hirata, K. Ishibashi, W. Sato et al., “Beta-mannosyl linkages inhibit CAWS arteritis by negatively regulating dectin-2-dependent signaling in spleen and dendritic cells,” Immunopharmacology and Immunotoxicology, vol. 35, pp. 594-604, 2013
  133. 133. B. Kerscher, J. A. Willment, and G. D. Brown, “The Dectin-2 family of C-type lectin-like receptors: an update,” International Immunology, vol. 25, no. 5, pp. 271-277, 2013
  134. 134. D. C. Ifrim, J. M. Bain,D. M. Reid et al., “The role of Dectin-2 for host defense against systemic infection with Candida glabrata,” Infection and Immunity, vol. 82, no. 3, pp. 1064-1073, 2014
  135. 135. L. Zhu, X. Zhao, C. Jiang et al., “C-type lectin receptors dectin-3 and dectin-2 form a heterodimeric pattern-recognition receptor for host defense against fungal infection,” Immunity, vol. 39, no. 2, pp. 324-334, 2013
  136. 136. A. Cambi, M. G. Netea, H. M. Mora-Montes et al., “Dendritic cell interaction with Candida albicans critically depends on Nlinked Mannan,” The Journal of Biological Chemistry, vol. 283, no. 29, pp. 20590-20599, 2008
  137. 137. J. S. Lam, H. Huang, and S. M. Levitz, “Effect of differential N-linked and O-linked mannosylation on recognition of fungal antigens by dendritic cells,” PLoS ONE, vol. 2, no. 10, Article ID e1009, 2007
  138. 138. P. R. Taylor, S. Gordon, and L. Martinez-Pomares, “The mannose receptor: linking homeostasis and immunity through sugar recognition,” Trends in Immunology, vol. 26, no. 2, pp. 104-110, 2005
  139. 139. U. Gazi,M. Rosas, S. Singh et al., “Fungal recognition enhances mannose receptor shedding through dectin-1 engagement,” Journal of Biological Chemistry, vol. 286, no. 10, pp. 7822-7829, 2011
  140. 140. F. L. van de Veerdonk, R. J. Marijnissen, B. J. Kullberg et al., “The macrophage mannose receptor induces IL-17 in response to Candida albicans,” Cell Host and Microbe, vol. 5, no. 4, pp. 329-340, 2009
  141. 141. S. E.M. Heinsbroek, P. R. Taylor, F. O.Martinez, L. Martinez- Pomares, G. D. Brown, and S. Gordon, “Stage-specific sampling by pattern recognition receptors during Candida albicans phagocytosis,” PLoS Pathogens, vol. 4, no. 11, Article ID e1000218, 2008
  142. 142. J. Zhang, S. D. Tachado, N. Patel et al., “Negative regulatory role of mannose receptors on human alveolar macrophage proinflammatory cytokine release in vitro,” Journal of Leukocyte Biology, vol. 78, no. 3, pp. 665-674, 2005
  143. 143. S. J. Lee,N. Zheng,M. Clavijo, andM.C.Nussenzweig, “Normal host defense during systemic candidiasis in mannose receptordeficient mice,” Infection and Immunity, vol. 71, no. 1, pp. 437-445, 2003
  144. 144. J. M. Dan, R. M. Kelly, C. K. Lee, and S. M. Levitz, “Role of the mannose receptor in a murine model of Cryptococcus neoformans infection,” Infection and Immunity, vol. 76, no. 6, pp. 2362-2367, 2008
  145. 145. F. Martinon, A. Mayor, and J. Tschopp, “The inflammasomes: guardians of the body,” Annual Review of Immunology, vol. 27, pp. 229-265, 2009
  146. 146. H. Kumar, T. Kawai, and S. Akira, “Pathogen recognition by the innate immune system,” International Reviews of Immunology, vol. 30, no. 1, pp. 16-34, 2011
  147. 147. C. Bryant and K. A. Fitzgerald, “Molecular mechanisms involved in inflammasome activation,” Trends in Cell Biology, vol. 19, no. 9, pp. 455-464, 2009
  148. 148. C. A. A. Van Der Graaf, M. G. Netea, B. Franke, S. E. Girardin, J.W. M. VanDerMeer, and B. J. Kullberg, “Nucleotide oligomerization domain 2 (Nod2) is not involved in the pattern recognition ofCandida albicans,” Clinical andVaccine Immunology, vol. 13, no. 3, pp. 423-425, 2006
  149. 149. S. Joly, N. Ma, J. J. Sadler, D. R. Soll, S. L. Cassel, and F. S. Sutterwala, “Cutting edge: Candida albicans hyphae formation triggers activation of the Nlrp3 inflammasome,” Journal of Immunology, vol. 183, no. 6, pp. 3578-3581, 2009
  150. 150. J. Tomalka, S. Ganesan, E. Azodi et al., “A novel role for the NLRC4 inflammasome in mucosal defenses against the fungal pathogen Candida albicans,” PLoS Pathogens, vol. 7, no. 12, Article ID e1002379, 2011
  151. 151. J. A. Kummer, R. Broekhuizen,H. Everett et al., “Inflammasome componentsNALP 1 and 3 showdistinct but separate expression profiles in human tissues suggesting a site-specific role in the inflammatory response,” Journal of Histochemistry and Cytochemistry, vol. 55, no. 5, pp. 443-452, 2007
  152. 152. Y.Mostefaoui, I.Claveau, andM. Rouabhia, “In vitro analyses of tissue structure and interleukin-1𝛽 expression and production by human oral mucosa in response to Candida albicans infections,” Cytokine, vol. 25, no. 4, pp. 162-171, 2004
  153. 153. M. Rouabhia, G. Ross, N. Pag’e, and J. Chakir, “Interleukin-18 and gamma interferon production by oral epithelial cells in response to exposure to Candida albicans or lipopolysaccharide stimulation,” Infection and Immunity, vol. 70, no. 12, pp. 7073-7080, 2002
  154. 154. G.Weindl, J. R. Naglik, S. Kaesler et al., “Human epithelial cells establish direct antifungal defense through TLR4-mediated signaling,” The Journal of Clinical Investigation, vol. 117, no. 12, pp. 3664-3672, 2007
  155. 155. F. Tardif, J. Goulet, A. Zakrazewski, P. Chauvin, and M. Rouabhia, “Involvement of interleukin-18 in the inflammatory response against oropharyngeal candidiasis,” Medical Science Monitor, vol. 10, no. 8, pp. BR239–BR249, 2004
  156. 156. O. Gross, H. Poeck, M. Bscheider et al., “Syk kinase signalling couples to the Nlrp3 inflammasome for anti-fungal host defence,” Nature, vol. 459, no. 7245, pp. 433-436, 2009
  157. 157. F. L. van de Veerdonk, B. J. Kullberg, J. W. van der Meer, N. A. Gow, and M. G. Netea, “Host-microbe interactions: innate pattern recognition of fungal pathogens,” Current Opinion in Microbiology, vol. 11, no. 4, pp. 305-312, 2008
  158. 158. A. G. Vonk,M.G.Netea, J.H.VanKrieken, Y. Iwakura, J.W. M. VanDerMeer, and B. J.Kullberg, “Endogenous interleukin (IL)-1𝛼 and IL-1𝛽 are crucial for host defense against disseminated candidiasis,” The Journal of Infectious Diseases, vol. 193, no. 10, pp. 1419-1426, 2006
  159. 159. A. G. Hise, J. Tomalka, S. Ganesan et al., “An essential role for the NLRP3 inflammasome in host defense against the human fungal pathogen Candida albicans,” Cell Host and Microbe, vol. 5, no. 5, pp. 487-497, 2009
  160. 160. M. H. Zaki,K.L.Boyd, P. Vogel,M. B. Kastan,M.Lamkanfi, and T. Kanneganti, “The NLRP3 Inflammasome Protects against Loss of Epithelial Integrity and Mortality during Experimental Colitis,” Immunity, vol. 32, no. 3, pp. 379-391, 2010
  161. 161. L. M. Rehaume, T. Jouault, andM. Chamaillard, “Lessons from the inflammasome: a molecular sentry linking Candida and Crohn’s disease,” Trends in Immunology, vol. 31, no. 5, pp. 171-175, 2010
  162. 162. A. Lev-Sagie, D. Prus, I. M. Linhares, Y. Lavy, W. J. Ledger, and S. S. Witkin, “Polymorphism in a gene coding for the inflammasome component NALP3 and recurrent vulvovaginal candidiasis in women with vulvar vestibulitis syndrome,” The American Journal of Obstetrics and Gynecology, vol. 200, no. 3, pp. 303.e1-303.e6, 2009
  163. 163. S. Yamasaki, M. Matsumoto, O. Takeuchi et al., “C-type lectin Mincle is an activating receptor for pathogenic fungus, Malassezia,” Proceedings of the National Academy of Sciences of the United States of America, vol. 106, no. 6, pp. 1897-1902, 2009
  164. 164. A. Bugarcic, K. Hitchens, A. G. Beckhouse, C. A. Wells, R. B. Ashman, and H. Blanchard, “Human and mouse macrophageinducible C-type lectin (Mincle) bind Candida albicans,” Glycobiology, vol. 18, no. 9, pp. 679-685, 2008
  165. 165. D. Vijayan, K. J. Radford, A. G. Beckhouse, R. B. Ashman, and C. A.Wells, “Mincle polarizes humanmonocyte and neutrophil responses to Candida albicans,” Immunology and Cell Biology, vol. 90, no. 9, pp. 889-895, 2012
  166. 166. M. Da Gl’oria Sousa, D. M. Reid, E. Schweighoffer et al., “Restoration of pattern recognition receptor costimulation to treat chromoblastomycosis, a chronic fungal infection of the skin,” Cell Host and Microbe, vol. 9, no. 5, pp. 436-443, 2011
  167. 167. W. Lee, J. Kang, J. Yan et al., “Neutrophils promote mycobacterial trehalose dimycolate-induced lung inflammation via the mincle pathway,” PLoS Pathogens, vol. 8, no. 4, Article ID e1002614, 2012
  168. 168. P. F. Zipfel, “Complement and immune defense: From innate immunity to human diseases,” Immunology Letters, vol. 126, no. 1-2, pp. 1-7, 2009
  169. 169. P. F. Zipfel and C. Skerka, “Complement, Candida, and cytokines: the role of C5a in host response to fungi,” European Journal of Immunology, vol. 42, no. 4, pp. 822-825, 2012
  170. 170. S. Luo, C. Skerka, O. Kurzai, and P. F. Zipfel, “Complement and innate immune evasion strategies of the human pathogenic fungus Candida albicans,“Molecular Immunology, vol. 56, no. 3, pp. 161-169, 2013
  171. 171. C. Speth, G. Rambach, R. W¨urzner, and C. Lass-Fl¨orl, “Complement and fungal pathogens: an update,” Mycoses, vol. 51, no. 6, pp. 477-496, 2008
  172. 172. N. Brouwer, K. M. Dolman, M. van Houdt, M. Sta, D. Roos, and T. W. Kuijpers, “Mannose-binding lectin (MBL) facilitates opsonophagocytosis of yeasts but not of bacteria despite MBL binding,” Journal of Immunology, vol. 180, no. 6, pp. 4124-4132, 2008
  173. 173. O. Neth, D. L. Jack, A. W. Dodds, H. Holzel, N. J. Klein, and M. W. Turner, “Mannose-binding lectin binds to a range of clinically relevant microorganisms and promotes complement deposition,” Infection and Immunity, vol. 68, no. 2, pp. 688-693, 2000
  174. 174. C. B. Forsyth and H. L. Mathews, “Lymphocytes utilize CD11b/CD18 for adhesion to Candida albicans,” Cellular Immunology, vol. 170, no. 1, pp. 91-100, 1996
  175. 175. T. Meri, A. Hartmann, D. Lenk et al., “The yeast Candida albicans binds complement regulators factor H and FHL-1,” Infection and Immunity, vol. 70, no. 9, pp. 5185-5192, 2002
  176. 176. T. Meri, A.M. Blom, A. Hartmann, D. Lenk, S. Meri, and P. F. Zipfel, “The hyphal and yeast forms of Candida albicans bind the complement regulator C4b-binding protein,” Infection and Immunity, vol. 72, no. 11, pp. 6633-6641, 2004
  177. 177. V. Agarwal, T. M. Asmat, S. Luo, I. Jensch, P. F. Zipfel, and S. Hammerschmidt, “Complement regulator factor H mediates a two-step uptake of Streptococcus pneumoniae by human cells,” The Journal of Biological Chemistry, vol. 285, no. 30, pp. 23486-23495, 2010
  178. 178. S. Poltermann, A. Kunert, M. Von Der Heide, R. Eck, A. Hartmann, and P. F. Zipfel, “Gpm1p is a factor H-, FHL-1-, and plasminogen-binding surface protein of Candida albicans,” Journal of Biological Chemistry, vol. 282, no. 52, pp. 37537-37544, 2007
  179. 179. S. Luo, S. Poltermann, A. Kunert, S. Rupp, and P. F. Zipfel, “Immune evasion of the human pathogenic yeast Candida albicans: Pra1 is a Factor H, FHL-1 and plasminogen binding surface protein,“Molecular Immunology, vol. 47,no. 2-3, pp. 541-550, 2009
  180. 180. S.Luo,A.M.Blom, S. Ruppet al., “ThepH-regulatedantigen1of Candida albicans binds the human complement inhibitor C4bbinding protein and mediates fungal complement evasion,” Journal of Biological Chemistry, vol. 286, no. 10, pp. 8021-8029, 2011
  181. 181. S. Luo, A. Hartmann, H. Dahse, C. Skerka, and P. F. Zipfel, “Secreted pH-regulated antigen 1 of Candida albicans blocks activation and conversion of complement C3,” Journal of Immunology, vol. 185, no. 4, pp. 2164-2173, 2010
  182. 182. R. B. Ashman, J. M. Papadimitriou, A. Fulurija et al., “Role of complement C5 and T lymphocytes in pathogenesis of disseminated and mucosal candidiasis in susceptible DBA/2 mice,” Microbial Pathogenesis, vol. 34, no. 2, pp. 103-113, 2003
  183. 183. R. B. Ashman, E. M. Bolitho, and J.M. Papadimitriou, “Patterns of resistance to Candida albicans in inbred mouse strains,” Immunology and Cell Biology, vol. 71, no. 3, pp. 221-225, 1993
  184. 184. R. B. Ashman, “Genetic determination of susceptibility and resistance in the pathogenesis of Candida albicans infection,” FEMS Immunology and Medical Microbiology, vol. 19, no. 3, pp. 183-189, 1997
  185. 185. I. Radovanovic, A. Mullick, and P. Gros, “Genetic control of susceptibility to infection with Candida albicans in mice,” PLoS ONE, vol. 6, no. 4,Article ID e18957, 2011
  186. 186. A. Mullick, M. Elias, S. Picard et al., “Dysregulated inflammatory response to Candida albicans in a C5-deficient mouse strain,” Infection and Immunity, vol. 72, no. 10, pp. 5868-5876, 2004
  187. 187. A. Mullick, Z. Leon, G. Min-Oo et al., “Cardiac failure in C5- deficient A/J mice after Candida albicans infection,” Infection and Immunity, vol. 74, no. 8, pp. 4439-4451, 2006
  188. 188. S.V. Tsoni, A.M. Kerrigan,M. J.Marakalala et al., “Complement C3 plays an essential role in the control of opportunistic fungal infections,” Infection and Immunity, vol. 77, no. 9, pp. 3679-3685, 2009
  189. 189. C. B. Forsyth and H. L. Mathews, “Lymphocyte adhesion to Candida albicans,” Infection and Immunity, vol. 70, no. 2, pp. 517-527, 2002
  190. 190. W. K. Ip and Y. L. Lau, “Role of mannose-binding lectin in the innate defense against Candida albicans: enhancement of complement activation, but lack of opsonic function, in phagocytosis by human dendritic cells,” Journal of Infectious Diseases, vol. 190, no. 3, pp. 632-640, 2004
  191. 191. M. C. Ghezzi, G. Raponi, S. Angeletti, and C.Mancini, “Serummediated enhancement of TNF-𝛼 release by human monocytes stimulated with the yeast form of Candida albicans,” Journal of Infectious Diseases, vol. 178, no. 6, pp. 1743-1749, 1998
  192. 192. A. Sonesson, L. Ringstad, E. Andersson Nordahl, M.Malmsten, M. M¨orgelin, and A. Schmidtchen, “Antifungal activity of C3a and C3a-derived peptides against Candida,” Biochimica et Biophysica Acta—Biomembranes, vol. 1768, no. 2, pp. 346-353, 2007
  193. 193. P. Miram’on, L. Kasper, and B. Hube, “Thriving within the host: candida spp. interactions with phagocytic cells,” Medical Microbiology and Immunology, vol. 202, no. 3, pp. 183-195, 2013
  194. 194. E. Svobodov’a, P. Staib, J. Losse, F. Hennicke, D. Barz, and M. J’ozsi, “Differential interaction of the two related fungal species Candida albicans and Candida dubliniensis with human neutrophils,” Journal of Immunology, vol. 189, no. 5, pp. 2502-2511, 2012
  195. 195. J. R. Linden, M. A. MacCani, S. S. Laforce-Nesbitt, and J. M Bliss, “High efficiency opsonin-independent phagocytosis of Candida parapsilosis by human neutrophils,” MedicalMycology, vol. 48, no. 2, pp. 355-364, 2010
  196. 196. W. L. Lee, R. E. Harrison, and S. Grinstein, “Phagocytosis by neutrophils,” Microbes and Infection, vol. 5, no. 14, pp. 1299-1306, 2003
  197. 197. B. Amulic, C. Cazalet, G. L. Hayes, K. D. Metzler, and A. Zychlinsky, “Neutrophil function: From mechanisms to disease,” Annual Review of Immunology, vol. 30, pp. 459-489, 2012
  198. 198. B. H. Segal, M. J. Grimm, A. N. H. Khan, W. Han, and T. S. Blackwell, “Regulation of innateimmunity byNADPHoxidase,” FreeRadical Biology andMedicine, vol. 53,no. 1, pp. 72-80, 2012
  199. 199. F. C. Fang, “Antimicrobial reactive oxygen and nitrogen species: concepts and controversies,” Nature Reviews Microbiology, vol. 2, no. 10, pp. 820-832, 2004
  200. 200. C. C. Winterbourn, M. B. Hampton, J. H. Livesey, and A. J. Kettle, “Modeling the reactions of superoxide and myeloperoxidase in the neutrophil phagosome: implications for microbial killing,” Journal of Biological Chemistry, vol. 281, no. 52, pp. 39860-39869, 2006
  201. 201. C. F. Urban, U. Reichard, V. Brinkmann, and A. Zychlinsky, “Neutrophil extracellular traps capture and kill Candida albicans and hyphal forms,” Cellular Microbiology, vol. 8, no. 4, pp. 668-676, 2006
  202. 202. C. F. Urban, D. Ermert, M. Schmid et al., “Neutrophil extracellular traps contain calprotectin, a cytosolic protein complex involved in host defense against Candida albicans,” PLoS Pathogens, vol. 5, no. 10, Article IDe1000639, 2009
  203. 203. K. Seider, A. Heyken, A. L¨uttich, P. Miram’on, and B. Hube, “Interaction of pathogenic yeasts with phagocytes: survival, persistence and escape,” Current Opinion in Microbiology, vol. 13, no. 4, pp. 392-400, 2010
  204. 204. P. Perumal, S. Mekala, C. Nombela, W. L. Chaffin, and C. Gil, “Proteomic analysis of cytoplasmic and surface proteins from yeast cells, hyphae, and biofilms of Candida albicans,” Proteomics, vol. 9, no. 8, pp. 2230-2252, 2009
  205. 205. F. O. Martinez, L. Helming, and S. Gordon, “Alternative activation of macrophages: an immunologic functional perspective,” Annual Review of Immunology, vol. 27, pp. 451-483, 2009
  206. 206. S. Gordon and F. O. Martinez, “Alternative activation of macrophages: mechanism and functions,” Immunity, vol. 32,no. 5, pp. 593-604, 2010
  207. 207. C. Jim’enez-L’opez and M. C. Lorenz, “Fungal immune evasion in a model host-pathogen interaction: Candida albicans versus macrophages,” PLoS Pathogens, vol. 9, Article IDe1003741, 2013
  208. 208. O. V. Vieira, R. J. Botelho, and S. Grinstein, “Phagosome maturation: aging gracefully,” Biochemical Journal, vol. 366, no. 3, pp. 689-704, 2002
  209. 209. D. M. Mosser and J. P. Edwards, “Exploring the full spectrum of macrophage activation,” Nature Reviews Immunology, vol. 8, no. 12, pp. 958-969, 2008
  210. 210. M. Stein, S. Keshav, N. Harris, and S. Gordon, “Interleukin 4 potently enhances murine macrophage mannose receptor activity: a marker of alternative immunologic macrophage activation,” Journal of ExperimentalMedicine, vol. 176, no. 1, pp. 287-292, 1992
  211. 211. M.Hesse,M.Modolell, A. C. La Flamme et al., “Differential regulation of nitric oxide synthase-2 and arginase-1 by type 1/type 2 cytokines in vivo: granulomatous pathology is shaped by the pattern of L-arginine metabolism,” Journal of Immunology, vol.167, no. 11, pp. 6533-6544, 2001
  212. 212. A. Tavanti, D. Campa, A. Bertozzi et al., “Candida albicans isolates with different genomic backgrounds display a differential response to macrophage infection,” Microbes and Infection, vol. 8, no. 3, pp. 791-800, 2006
  213. 213. I. E. Frohner, C. Bourgeois, K. Yatsyk,O.Majer, andK. Kuchler, “Candida albicans cell surface superoxide dismutases degrade host-derived reactive oxygen species to escape innate immune surveillance,“MolecularMicrobiology, vol. 71,no. 1, pp. 240-252, 2009
  214. 214. B. D. Ullmann, H. Myers, W. Chiranand et al., “Inducible defense mechanism against nitric oxide in Candida albicans,” Eukaryotic Cell, vol. 3, no. 3, pp. 715-723, 2004
  215. 215. E. Fern’andez-Arenas, V. Cabez’on, C. Bermejo et al., “Integrated proteomics and genomics strategies bring new insight into Candida albicans response upon macrophage interaction,” Molecular and Cellular Proteomics, vol. 6, no. 3, pp. 460-478, 2007

Written By

Snigdha Pattnaik, Laxmidhar Maharana and Manoj Sethi

Submitted: 17 November 2020 Reviewed: 30 July 2021 Published: 05 September 2021