2
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Calcineurin: The Achilles’ heel of fungal pathogens

      review-article
      * , , * ,
      PLOS Pathogens
      Public Library of Science

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          What is calcineurin? Calcineurin is a highly conserved serine-threonine specific protein phosphatase and was named based on its discovery as a calcium- and calmodulin-binding protein that is highly abundant in the central nervous system (calci+neurin) [1]. Calcineurin is a heterodimer comprised of a catalytic A subunit (CNA) and a regulatory B subunit (CNB) [2,3]. In the basal state, CNB is bound by 2 Ca2+ ions and calcineurin is kept inactive through an autoinhibitory domain (AID) that loops into the active site of CNA [2] (Fig 1). Upon calcium influx, CNB binds to 2 additional Ca2+ ions causing structural changes in the CNA-CNB heterodimer. Finally, the heterodimer is associated with another calcium-binding protein, calmodulin, which itself is primed via structural changes through binding of 4 Ca2+ ions. Calmodulin binding triggers the release of the AID from the catalytic site of the enzyme rendering calcineurin fully active. Once activated, calcineurin binds to its substrates by docking onto 2 short-linear motifs, PxIxIT and LxVP, and dephosphorylates substrates to govern their subcellular localization and functions [3]. 10.1371/journal.ppat.1011445.g001 Fig 1 Calcineurin signaling in fungal pathogens. Calcineurin signaling is initiated upon calcium influx and binding of calcium-calmodulin (CaM) to the calcineurin complex (CNA and CNB). The binding of CaM releases an autoinhibitory domain that occupies the active site in the catalytic subunit CNA converting calcineurin from an inactive “OFF” configuration to a fully active “ON” state. Upon activation, calcineurin dephosphorylates its substrates, which include a conserved transcription factor, Crz1, and several others that remain unidentified in most fungal species. Through these targets, calcineurin governs stress responses, morphological transitions, and virulence in fungal pathogens of humans and plants. As described in the right-side panel, calcineurin directs yeast–hyphal dimorphic transitions in some fungi, whereas it is required for vegetative yeast or hyphal growth in other fungal species. In some cases, calcineurin orchestrates the development of specialized structures, such as formation of appressoria in M. oryzae, that are required for infection and virulence. Calcineurin activity can be inhibited by FK506 and cyclosporine (CsA), both of which are immunosuppressive drugs when bound to FKBP12 or cyclophilin A (Cyp). The figure was created with BioRender.com. How important is calcineurin for fungal pathogens? Calcineurin plays key roles in fungi in response to calcium influx and is the only known calcium-responsive phosphatase. Calcineurin has been studied in several fungal pathogens of both humans and plants and plays crucial roles in stress adaptation and pathogenicity (Fig 1 and Table 1). In the human fungal pathogen, Candida albicans, calcineurin is required for survival in serum and plays an important role during hyphal growth required to cause infections [4–6]. Additionally, loss of function or inhibition of calcineurin also results in enhanced susceptibility to antifungal drugs in widespread clinical use such as fluconazole. Calcineurin plays a crucial role in the hyphal growth and virulence of Aspergillus fumigatus, a filamentous fungal pathogen [7]. The catalytic subunit, CNA, localizes to the active sites of hyphal growth and septum formation, playing an important role in growth and septation in A. fumigatus [8]. Inhibiting calcineurin activity also results in hypersensitivity to other cell wall inhibitors in this fungus suggesting a major role for calcineurin in proper cell wall synthesis and repair [9]. 10.1371/journal.ppat.1011445.t001 Table 1 Calcineurin functions in fungal pathogens. Fungal pathogen Calcineurin functions References Human pathogens Aspergillus fumigatus Hyphal growth, cell wall integrity, septation, cation homeostasis, antifungal drug resistance, and virulence [7–9,22] Candida albicans Hyphal growth, ER stress response, cell wall integrity, azole tolerance, growth in serum, and virulence [4–6,20,34,37] Candida glabrata Thermotolerance, ER stress response, cell wall integrity, azole tolerance, growth in serum, and virulence [38,39] Cryptococcus neoformans Thermotolerance, cell wall integrity, antifungal drug tolerance, postmating dimorphic transition from yeast to hyphae, and virulence [10,11,21,24] Mucor circinelloides Vegetative dimorphic transition from yeast to hyphae, hyphal growth, antifungal drug resistance, and virulence [14,40] Paracoccidioides brasiliensis Vegetative dimorphic transition from hyphae to yeast, yeast and hyphal growth, and calcium homeostasis [13] Talaromyces marneffei Hyphal growth, conidiation and conidia germination, cell wall integrity, osmotic stress response, survival in macrophages, and virulence [16] Trichosporon asahii Thermotolerance, cell wall integrity, ER stress response, hyphal formation, and virulence [15] Plant pathogens Botrytis cinerea Conidiation, cation homeostasis, cell wall integrity, and virulence [41] Magnaporthe oryzae Mycelial growth, conidiation, appressorium formation, and virulence [17,23,42] Ustilago maydis Postmating dimorphic transition from yeast to hyphae, and virulence [18] Ustilago hordei Thermotolerance, cell wall integrity, cation homeostasis, pH stress, and virulence [43] Functions listed in bold indicate roles that are known to be either completely or partially independent of Crz1. In Cryptococcus neoformans, mutation or inhibition of calcineurin renders cells inviable at 37°C, and avirulent in mice [10]. In addition, calcineurin is also required for hyphal growth in C. neoformans, which is essential for completing the sexual cycle and producing infectious spores [11]. Microscopy studies demonstrated that calcineurin re-localizes to septation sites as well as processing-bodies (P-bodies) and stress granules during 37°C heat stress [12]. Calcineurin has also been found to play essential roles in virulence in several other human fungal pathogens including Mucor circinelloides, Paracoccidioides brasiliensis, Talaromyces marneffei, and Trichosporon asahii [13–16]. Magnaporthe oryzae is a serious threat to global food security and is responsible for approximately a 30% loss in rice productivity each year. M. oryzae infects plant leaves through specialized structures called appressoria. Calcineurin is required for the formation of appressoria and therefore, it is essential for pathogenesis [17]. Calcineurin also plays a critical role in the pathogenicity of Ustilago maydis, a plant fungal pathogen that infects corn [18]. U. maydis undergoes a postmating morphological transition from yeast to hyphal growth that is essential for virulence. Mutation of calcineurin results in the production of multi-budded yeast cells that fail to form hyphae and are thus unable to cause infections. Overall, calcineurin directs stress responses and morphological changes in each of these pathogens and thus serves as a globally conserved virulence factor. What are calcineurin’s substrates? One of the most conserved calcineurin targets in fungi is Crz1, a zinc-finger transcription factor, which is a homolog of NFAT in humans [19]. Upon dephosphorylation by activated calcineurin, Crz1 translocates into the nucleus and regulates the transcription of target genes involved in stress responses. While Crz1 is known to perform roles in the calcineurin pathway in several fungal species, its activity fails to account for all of calcineurin’s functions. For example, Crz1 in C. albicans plays roles in pH sensitivity, hyphal growth, and drug tolerance but is not required for virulence, unlike calcineurin [20]. Similarly, Crz1 deletion does not lead to the same level of thermal sensitivity as deletion or inhibition of calcineurin in C. neoformans [21]. In addition, Crz1 is not required for hyphal growth and sporulation in this species, in contrast to calcineurin. While both calcineurin and the Crz1 homolog CrzA play an important role in the virulence of A. fumigatus, CrzA deletion does not phenocopy the severe hyphal growth defect of the calcineurin mutant, and accordingly, calcineurin plays a more prominent role in cell wall biosynthesis compared to the more restricted role of Crz1 [22]. Interestingly, the Crz1 homolog in M. oryzae is not required for appressorium formation [23], but Crz1 mutation still reduces rice infection rates. Crz1 in U. maydis remains to be identified. Overall, these studies demonstrate that Crz1 is a conserved and key downstream effector of calcineurin in most fungal pathogens, but also emphasizes that additional substrates of calcineurin must also play critical roles in stress responses. Surprisingly, aside from Crz1, most calcineurin targets in pathogenic fungi remain understudied and poorly characterized, except for some potential candidates identified in C. neoformans. A phosphoproteome study in this fungus identified several calcineurin targets, in addition to Crz1 [24]. Many of these substrates localize to P-bodies/stress granules and are predicted to play important roles in RNA processing. Additionally, substrates involved in septation and vesicle trafficking were also identified, in accord with previous localization studies [12,25]. Analysis of calcineurin substrates revealed that both Crz1 and P-body/stress granule targets contribute to thermal stress adaptation and virulence but their mutation confers only intermediate phenotypes in comparison to wild-type and calcineurin mutants [24]. While Crz1 controls the expression of several cell wall biosynthesis genes in a calcineurin-dependent manner, thus regulating cell wall integrity, the roles of calcineurin in P-bodies/stress granules are not well understood but may involve RNA metabolism. Interestingly, combined mutation of Crz1 and P-body targets does not recapitulate the phenotypes of calcineurin deletion, suggesting that additional substrates may be important for calcineurin functions [24]. A recent study revealed that Crz1 also localizes to stress granules and septa upon heat stress, similar to calcineurin [26]. Interestingly, the study found that Crz1 localizes to stress granules prior to its translocation to the nucleus hinting at the possibility that calcineurin might be dephosphorylating Crz1 at stress granules. Can calcineurin serve as a drug target? Calcineurin, due to its diverse and critical roles, is an attractive antifungal drug target candidate. Due to its stimulatory role in human T-cells, it is an established target of 2 immunosuppressive drugs, FK506 and cyclosporine (CsA), both of which inhibit calcineurin as drug–protein complexes after binding to their respective immunophilin targets, FKBP12, and cyclophilin A [2]. Both FK506 and CsA also exhibit broad antifungal activity highlighting the potential of calcineurin as an antifungal drug target [27]. However, the conservation of calcineurin signaling between humans and fungi requires alternative approaches to identify aspects for fungal-specific targeting. Recently, structure-guided synthetic analogs of FK506 have been developed that demonstrate greater relative specificity for fungal calcineurin than its human counterpart [28–30]. Strengthening this proof-of-concept, these analogs retain antifungal activity with reduced immunosuppressive activity signifying that this approach could yield drug candidate leads. Such an approach of structure-guided synthesis of inhibitors can also be applied to target downstream components of calcineurin, such as Crz1. Another approach could be to identify unique structural and/or sequence features that differ between fungi and the host for drug targeting. Interestingly, a filamentous fungal-specific phosphorylated serine proline-rich (SPRR) domain was identified in A. fumigatus CNA homolog, which is important for regular hyphal growth, revealing unique protein sequence features that could be exploited for drug development [31]. These findings have revealed a hidden potential of calcineurin inhibitors and suggest more extensive research involving structure-guided drug design is warranted. What remains to be learned about calcineurin in pathogenic fungi? Fungal infections pose a significant challenge to both global human health and food security [32,33]. This threat is further magnified by the increasing population of immunocompromised people and increasing resistance of fungal pathogens to existing treatment options. This scenario suggests that more research is needed to identify novel fungal-specific drug targets that could lead to the development of fungicidal drugs. Studying and characterizing calcineurin in detail in key fungal pathogens has the potential to identify fungal-specific drug targets within this signaling network. However, at present, there is a substantial gap between our knowledge of calcineurin’s roles in fungi and how these are performed at a molecular level. Most research has focused on the functions of calcineurin in cell wall integrity pathways and studying the contributions of Crz1 as a downstream effector. While both of these factors are important and necessary components of the signaling network, they are not sufficient to fully explain the role of calcineurin in fungal pathogenesis. Therefore, more focused and mechanistic approaches are required to critically dissect the roles of calcineurin and its substrates to provide important breakthroughs. Future studies should delineate a more complete repertoire of calcineurin substrates in fungi. This may result in the identification of calcineurin effectors that are required for virulence but might be specific to a fungal species. In addition, characterizing these effectors in multiple species could also reveal a set of substrates or functions that are specific to fungi and absent from humans. For example, calcineurin interactions at septa appear to be conserved across fungi and thus reflect interactions with fungal-specific targets [8,12]. In addition, such studies may also reveal the basis of the requirement for calcineurin in specific host niches. For example, the requirement for calcineurin in C. albicans virulence in mice depends on the mode of infection, where it is essential for systemic infection, but not for pulmonary or vaginal infection [4,5,34]. Such studies could help delineate specific host factors that contribute to the establishment of infections revealing interesting infectious disease biology involving calcineurin signaling. This might include delineating roles of calcineurin effectors in different host niches allowing the development of effector-targeting drugs that might be condition specific. Another exciting aspect of calcineurin signaling that is understudied is the documentation of kinases that phosphorylate calcineurin substrates and thereby regulate their function. For example, the kinase responsible for phosphorylating Crz1 and required for its nuclear export in fungal pathogens remains uncharacterized. Interestingly, 2 separate kinases, Hrr25, and protein kinase A, are known to phosphorylate Crz1 in S. cerevisiae suggesting that there may be more than 1 Crz1-regulating kinase in other fungi as well [19,35]. Calmodulin is known to also activate several kinases in response to calcium signaling [36] and some of these kinases may phosphorylate calcineurin’s substrates. Recognizing and studying these negative regulators of calcineurin signaling will be important and may lead to the identification of core calcineurin effectors that play essential roles in fungal pathogenesis. Calcineurin plays a fundamental role in virtually every fungal pathogen studied to date and yet research into this pathway can be accelerated. Going forward, further research into calcineurin signaling will be necessary to fully exploit the therapeutic potential of this essential virulence network. Targeting calcium–calcineurin signaling has the potential to significantly influence not only human health and global food resources but also biodiversity because of its significance in a broad range of fungal pathogens. As a true Achilles’ heel, calcineurin signaling can be aimed at and targeted to counter the emerging threats of fungal diseases.

          Related collections

          Most cited references42

          • Record: found
          • Abstract: found
          • Article: not found

          Calcineurin is required for virulence of Cryptococcus neoformans.

          Cyclosporin A (CsA) and FK506 are antimicrobial, immunosuppressive natural products that inhibit signal transduction. In T cells and Saccharomyces cerevisiae, CsA and FK506 bind to the immunophilins cyclophilin A and FKBP12 and the resulting complexes inhibit the Ca2+-regulated protein phosphatase calcineurin. We find that growth of the opportunistic fungal pathogen Cryptococcus neoformans is sensitive to CsA and FK506 at 37 degrees C but not at 24 degrees C, suggesting that CsA and FK506 inhibit a protein required for C. neoformans growth at elevated temperature. Genetic evidence supports a model in which immunophilin-drug complexes inhibit calcineurin to prevent growth at 37 degrees C. The gene encoding the C. neoformans calcineurin A catalytic subunit was cloned and disrupted by homologous recombination. Calcineurin mutant strains are viable but do not survive in vitro conditions that mimic the host environment (elevated temperature, 5% CO2 or alkaline pH) and are no longer pathogenic in an animal model of cryptococcal meningitis. Introduction of the wild-type calcineurin A gene complemented these growth defects and restored virulence. Our findings demonstrate that calcineurin is required for C. neoformans virulence and may define signal transduction elements required for fungal pathogenesis that could be targets for therapeutic intervention.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Calcineurin: a calcium- and calmodulin-binding protein of the nervous system.

            The inhibitory protein that binds calmodulin and thus prevents activation of several Ca2+-dependent enzymes by calmodulin is shown to also bind four Ca2+ per mol of protein with high affinity (Kd less than or equal to 10(-6) M). On the basis of its Ca2+- binding properties and its localization to nervous tissue, the inhibitory protein is now called "calcineurin." Calcineurin is composed of two subunits: calcineurin A (61,000 Mr) which interacts with calmodulin in a Ca2+-dependent fashion, and calcineurin B (15,000 Mr) which binds Ca2+. The interaction of calcineurin A with calcineurin B is independent of Ca2+ or Mg2+. The dual interaction of calcineurin A with two different Ca2+-binding components and the high affinity of calcineurin for Ca2+ suggest a possible role for calcineurin in the regulation of free Ca2+ concentrations in the nervous system. Calcineurin may thereby modulate the release and action of neurotransmitters.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Calcineurin controls growth, morphology, and pathogenicity in Aspergillus fumigatus.

              Calcineurin is implicated in a myriad of human diseases as well as homeostasis and virulence in several major human pathogenic microorganisms. The fungus Aspergillus fumigatus is a leading cause of infectious death in the rapidly expanding immunocompromised patient population. Current antifungal treatments for invasive aspergillosis are often ineffective, and novel therapeutic approaches are urgently needed. We demonstrate that a mutant of A. fumigatus lacking the calcineurin A (cnaA) catalytic subunit exhibited defective hyphal morphology related to apical extension and polarized growth, which resulted in drastically decreased filamentation. The delta cnaA mutant lacked the extensive lattice of invading hyphae seen with the wild-type and complemented strains. Sporulation was also affected in the delta cnaA mutant, including morphological conidial defects with the absence of surface rodlets and the added presence of disjunctors creating long conidial chains. Infection with the delta cnaA mutant in several distinct animal models with different types of immunosuppression and inoculum delivery led to a profound attenuation of pathogenicity compared to infection with the wild-type and complemented strains. Lung tissue from animals infected with the delta cnaA mutant showed a complete absence of hyphae, in contrast to tissue from animals infected with the wild-type and complemented strains. Quantitative fungal burden and pulmonary infarct scoring confirmed these findings. Our results support the clinical observation that substantially decreasing fungal growth can prevent disease establishment and decrease mortality. Our findings reveal that calcineurin appears to play a globally conserved role in the virulence of several pathogenic fungi and yet plays specialized roles in each and can be an excellent target for therapeutic intervention.
                Bookmark

                Author and article information

                Contributors
                Role: ConceptualizationRole: Funding acquisitionRole: VisualizationRole: Writing – original draftRole: Writing – review & editing
                Role: ConceptualizationRole: Funding acquisitionRole: SupervisionRole: Writing – review & editing
                Role: Editor
                Journal
                PLoS Pathog
                PLoS Pathog
                plos
                PLOS Pathogens
                Public Library of Science (San Francisco, CA USA )
                1553-7366
                1553-7374
                6 July 2023
                July 2023
                : 19
                : 7
                : e1011445
                Affiliations
                [001] Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
                University of Melbourne, AUSTRALIA
                Author notes

                The authors have declared that no competing interests exist.

                Author information
                https://orcid.org/0000-0003-2650-9035
                Article
                PPATHOGENS-D-23-00555
                10.1371/journal.ppat.1011445
                10325075
                37410706
                c176032f-d2b3-4d21-b616-b3b63849d460
                © 2023 Yadav, Heitman

                This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

                History
                Page count
                Figures: 1, Tables: 1, Pages: 8
                Funding
                Funded by: funder-id http://dx.doi.org/10.13039/100015691, Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases;
                Award ID: R01-AI039115-25
                Award Recipient :
                Funded by: funder-id http://dx.doi.org/10.13039/100015691, Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases;
                Award ID: R01-AI050113-18
                Award Recipient :
                Funded by: funder-id http://dx.doi.org/10.13039/100015691, Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases;
                Award ID: R01-AI172451-01
                Award Recipient :
                Funded by: funder-id http://dx.doi.org/10.13039/100006967, Center for AIDS Research, Duke University;
                Award ID: Pilot grant through 5P30 AI064518
                Award Recipient :
                This work was supported by NIH/NIAID R01 award AI039115-25, R01 grant AI050113-18, and AI172451-01 awarded to JH. JH is also Co-Director and Fellow of the CIFAR program Fungal Kingdom: Threats & Opportunities. This work was funded in part by a 2022 developmental grant to VY from the Duke University Center for AIDS Research (CFAR), an NIH funded program (5P30 AI064518). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
                Categories
                Pearls
                Biology and Life Sciences
                Microbiology
                Medical Microbiology
                Microbial Pathogens
                Fungal Pathogens
                Medicine and Health Sciences
                Pathology and Laboratory Medicine
                Pathogens
                Microbial Pathogens
                Fungal Pathogens
                Biology and Life Sciences
                Mycology
                Fungal Pathogens
                Biology and Life Sciences
                Organisms
                Eukaryota
                Fungi
                Biology and Life Sciences
                Plant Science
                Plant Pathology
                Plant Pathogens
                Plant Fungal Pathogens
                Biology and Life Sciences
                Cell Biology
                Cellular Structures and Organelles
                Cell Walls
                Plant Cell Walls
                Biology and Life Sciences
                Cell Biology
                Cellular Types
                Plant Cells
                Plant Cell Walls
                Biology and Life Sciences
                Cell Biology
                Plant Cell Biology
                Plant Cells
                Plant Cell Walls
                Biology and Life Sciences
                Plant Science
                Plant Cell Biology
                Plant Cells
                Plant Cell Walls
                Biology and Life Sciences
                Organisms
                Eukaryota
                Fungi
                Yeast
                Candida
                Candida Albicans
                Biology and Life Sciences
                Microbiology
                Medical Microbiology
                Microbial Pathogens
                Fungal Pathogens
                Candida Albicans
                Medicine and Health Sciences
                Pathology and Laboratory Medicine
                Pathogens
                Microbial Pathogens
                Fungal Pathogens
                Candida Albicans
                Biology and Life Sciences
                Mycology
                Fungal Pathogens
                Candida Albicans
                Research and Analysis Methods
                Animal Studies
                Experimental Organism Systems
                Yeast and Fungal Models
                Candida Albicans
                Biology and Life Sciences
                Mycology
                Fungal Structure
                Biology and Life Sciences
                Plant Science
                Plant Pathology
                Plant Pathogens
                Biology and Life Sciences
                Cell Biology
                Signal Transduction
                Cell Signaling
                Signaling Cascades
                Calcineurin Signaling Cascade

                Infectious disease & Microbiology
                Infectious disease & Microbiology

                Comments

                Comment on this article