1
|
Kuo CF, Chen YY, Chiu CC, Chiu CW, Li TC, Chang YS, Tsao N. Comparative in vitro efficacy of AR-12 derivatives against Streptococcus pyogenes. J Antimicrob Chemother 2024:dkae462. [PMID: 39704166 DOI: 10.1093/jac/dkae462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024] Open
Abstract
OBJECTIVES Group A Streptococcus (GAS) results in invasive diseases. Our published studies show that AR-12 can directly kill GAS. However, AR-12 is toxic to the human microvascular endothelial cells (HMEC-1 cells) even at its MIC. In this study, we examined various AR-12 pyrrole derivatives, selected the most effective one and used it to combat GAS. METHODS The bacterial numbers after treatment with AR-12 derivatives were assessed using either spectrophotometry or the colony-forming unit assay. The integrity of cell envelope and the contents of proteins and nucleic acids in GAS were sequentially examined by staining with SYTOX Green, SYPRO or propidium iodide. The protein expression was assessed by western blotting. The cytotoxicity of AR-12 derivatives was evaluated using WST-1 assay, the lactate dehydrogenase release assay and Annexin V staining. RESULTS We tested AR-12 pyrrole derivatives P12, P12-3 and P12-8 on GAS growth and found that P12 and P12-8 were effective against various M-type strains. Both P12 and P12-8 disrupted the GAS envelope and reduced protein and nucleic acid content in GAS at their MICs. At sub-MIC levels, both P12 and P12-8 inhibited GAS chaperone protein and streptococcal pyrogenic exotoxin B expression. P12 and P12-8 also exhibited a synergistic effect with gentamicin against GAS. However, only P12-8 did not affect cell death at its MIC. Besides its bactericidal efficacy, P12-8 also enhanced the clearance of intracellular bacteria in GAS-infected A549 and HMEC-1 cells. CONCLUSIONS Among these three AR-12 derivatives, P12-8 had the best potential to be an alternative agent to fight against GAS.
Collapse
Affiliation(s)
- Chih-Feng Kuo
- School of Medicine, I-Shou University, Kaohsiung City, Taiwan
- Department of Nursing, I-Shou University, Kaohsiung City, Taiwan
| | - You-Yan Chen
- Department of Medical Laboratory Science, I-Shou University, Kaohsiung City, Taiwan
| | - Ching-Chen Chiu
- Department of Medical Laboratory Science, I-Shou University, Kaohsiung City, Taiwan
| | - Chih-Wei Chiu
- Department of Medical Laboratory Science, I-Shou University, Kaohsiung City, Taiwan
| | - Tang-Chi Li
- Department of Medical Laboratory Science, I-Shou University, Kaohsiung City, Taiwan
| | - Yu-Shan Chang
- Department of Laboratory Medicine, E-DA Hospital, Kaohsiung City, Taiwan
| | - Nina Tsao
- Department of Medical Laboratory Science, I-Shou University, Kaohsiung City, Taiwan
| |
Collapse
|
2
|
Cui X, Wang L, Lü Y, Yue C. Development and research progress of anti-drug resistant fungal drugs. J Infect Public Health 2022; 15:986-1000. [PMID: 35981408 DOI: 10.1016/j.jiph.2022.08.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 08/01/2022] [Accepted: 08/05/2022] [Indexed: 11/24/2022] Open
Abstract
With the widespread use of immunosuppressive agents and the increase in patients with severe infections, the incidence of fungal infections worldwide has increased year by year. The fungal pathogens Candida albicans, Cryptococcus neoformans and Aspergillus fumigatus cause a total of more than 1 million deaths each year. Long-term use of antifungal drugs can easily lead to fungal resistance, and the prevalence of drug-resistant fungi is a major global health challenge. In order to effectively control global fungal infections, there is an urgent need for new drugs that can exert effective antifungal activity and overcome drug resistance. We must promote the discovery of new antifungal targets and drugs, and find effective ways to control drug-resistant fungi through different ways, so as to reduce the threat of drug-resistant fungi to human life, health and safety. In the past few years, certain progress has been made in the research and development of antifungal drugs. In addition to summarizing some of the antifungal drugs currently approved by the FDA, this review also focuses on potential antifungal drugs, the repositioned drugs, and drugs that can treat drug-resistant bacteria and fungal infections, and provide new ideas for the development of antifungal drugs in the future.
Collapse
Affiliation(s)
- Xiangyi Cui
- Key Laboratory of Microbial Drugs Innovation and Transformation of Yan'an, School of Basic Medicine, Yan'an University, Yan'an 716000, Shaanxi, China; Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources,Yan'an University, NO.580 Shengdi Road, Baota District, Yan'an 716000, Shaanxi, China.
| | - Lanlin Wang
- Key Laboratory of Microbial Drugs Innovation and Transformation of Yan'an, School of Basic Medicine, Yan'an University, Yan'an 716000, Shaanxi, China; Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources,Yan'an University, NO.580 Shengdi Road, Baota District, Yan'an 716000, Shaanxi, China.
| | - Yuhong Lü
- Key Laboratory of Microbial Drugs Innovation and Transformation of Yan'an, School of Basic Medicine, Yan'an University, Yan'an 716000, Shaanxi, China; Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources,Yan'an University, NO.580 Shengdi Road, Baota District, Yan'an 716000, Shaanxi, China.
| | - Changwu Yue
- Key Laboratory of Microbial Drugs Innovation and Transformation of Yan'an, School of Basic Medicine, Yan'an University, Yan'an 716000, Shaanxi, China; Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources,Yan'an University, NO.580 Shengdi Road, Baota District, Yan'an 716000, Shaanxi, China.
| |
Collapse
|
3
|
Bhattacharya S, Oliveira NK, Savitt AG, Silva VKA, Krausert RB, Ghebrehiwet B, Fries BC. Low Glucose Mediated Fluconazole Tolerance in Cryptococcus neoformans. J Fungi (Basel) 2021; 7:jof7060489. [PMID: 34207384 PMCID: PMC8233753 DOI: 10.3390/jof7060489] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/13/2021] [Accepted: 06/16/2021] [Indexed: 01/07/2023] Open
Abstract
Chronic meningoencephalitis is caused by Cryptococcus neoformans and is treated in many parts of the world with fluconazole (FLC) monotherapy, which is associated with treatment failure and poor outcome. In the host, C. neoformans propagates predominantly under low glucose growth conditions. We investigated whether low glucose, mimicked by growing in synthetic media (SM) with 0.05% glucose (SMlowglu), affects FLC-resistance. A > 4-fold increase in FLC tolerance was observed in seven C. neoformans strains when minimum inhibitory concentration (MIC) was determined in SMlowglu compared to MIC in SM with normal (2%) glucose (SMnlglu). In SMlowglu, C. neoformans cells exhibited upregulation of efflux pump genes AFR1 (8.7-fold) and AFR2 (2.5-fold), as well as decreased accumulation (2.6-fold) of Nile Red, an efflux pump substrate. Elevated intracellular ATP levels (3.2-fold and 3.4-fold), as well as decreased mitochondrial reactive oxygen species levels (12.8-fold and 17-fold), were found in the presence and absence of FLC, indicating that low glucose altered mitochondrial function. Fluorescence microscopy revealed that mitochondria of C. neoformans grown in SMlowglu were fragmented, whereas normal glucose promoted a reticular network of mitochondria. Although mitochondrial membrane potential (MMP) was not markedly affected in SMlowglu, it significantly decreased in the presence of FLC (12.5-fold) in SMnlglu, but remained stable in SMlowglu-growing C. neoformans cells. Our data demonstrate that increased FLC tolerance in low glucose-growing C. neoformans is the result of increased efflux pump activities and altered mitochondrial function, which is more preserved in SMlowglu. This mechanism of resistance is different from FLC heteroresistance, which is associated with aneuploidy of chromosome 1 (Chr1).
Collapse
Affiliation(s)
- Somanon Bhattacharya
- Division of Infectious Diseases, Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (S.B.); (V.K.A.S.)
| | - Natalia Kronbauer Oliveira
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (N.K.O.); (A.G.S.); (R.B.K.)
| | - Anne G. Savitt
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (N.K.O.); (A.G.S.); (R.B.K.)
| | - Vanessa K. A. Silva
- Division of Infectious Diseases, Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (S.B.); (V.K.A.S.)
| | - Rachel B. Krausert
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (N.K.O.); (A.G.S.); (R.B.K.)
| | - Berhane Ghebrehiwet
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA;
| | - Bettina C. Fries
- Division of Infectious Diseases, Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (S.B.); (V.K.A.S.)
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (N.K.O.); (A.G.S.); (R.B.K.)
- Veterans Administration Medical Center, Northport, NY 11768, USA
- Correspondence:
| |
Collapse
|
4
|
Teymuri M, Shams-Ghahfarokhi M, Razzaghi-Abyaneh M. Inhibitory effects and mechanism of antifungal action of the natural cyclic depsipeptide, aureobasidin A against Cryptococcus neoformans. Bioorg Med Chem Lett 2021; 41:128013. [PMID: 33811994 DOI: 10.1016/j.bmcl.2021.128013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/22/2021] [Accepted: 03/28/2021] [Indexed: 02/08/2023]
Abstract
Cryptococcosis is an opportunistic fungal infection caused mainly by Cryptococcus neoformans. The aim of the present study was to evaluate the inhibitory effect of aureobasidin A on C. neoformans with special focus on its mode of action. The effect of aureobasidin A on cell membrane ergosterol content, cell wall permeability, membrane pumps activities, the total oxidant status (TOS) and melanin production was evaluated. Cytotoxicity and cell hemolysis, and laccase (LacI) and β1,2-xylosyltransferase (Cxt1p) gene expression were also evaluated. Aureobasidin A reduced melanin production and increased extracellular potassium leakage at 0.5 × MIC concentration. This peptide has no effect on fungal cell wall integrity. Cell membrane ergosterol content was decreased by 29.1% and 41.8% at 0.5 × MIC and 1 × MIC concentrations (2 and 4 µL/mL) in aureobasidin A treated samples, respectively. TOS level was significantly increased without activation of antioxidant enzymes. Lac1 gene was over-expressed (11.7-fold), while Cxt1p gene was down regulated (0.2-fold) following treatment with aureobasidin A. Overall, our results indicated that aureobasidin A inhibits C. neoformans growth by targeting different sites in fungal cells and it may be considered as a promising compound to use as an antifungal in treatment of clinical cryptococcosis.
Collapse
Affiliation(s)
- Mostafa Teymuri
- Department of Mycology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14115-331, Iran
| | - Masoomeh Shams-Ghahfarokhi
- Department of Mycology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14115-331, Iran.
| | | |
Collapse
|
5
|
Gaylord EA, Choy HL, Doering TL. Dangerous Liaisons: Interactions of Cryptococcus neoformans with Host Phagocytes. Pathogens 2020; 9:E891. [PMID: 33121050 PMCID: PMC7692806 DOI: 10.3390/pathogens9110891] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 10/22/2020] [Accepted: 10/25/2020] [Indexed: 02/07/2023] Open
Abstract
Cryptococcus neoformans is an opportunistic fungal pathogen and a leading cause of death in immunocompromised individuals. The interactions of this yeast with host phagocytes are critical to disease outcome, and C. neoformans is equipped with an array of factors to modulate these processes. Cryptococcal infection begins with the deposition of infectious particles into the lungs, where the fungal cells deploy various antiphagocytic factors to resist internalization by host cells. If the cryptococci are still engulfed, they can survive and proliferate within host cells by modulating the phagolysosome environment in which they reside. Lastly, cryptococcal cells may escape from phagocytes by host cell lysis, nonlytic exocytosis, or lateral cell-to-cell transfer. The interactions between C. neoformans and host phagocytes also influence the dissemination of this pathogen to the brain, where it may cross the blood-brain barrier and cause an often-fatal meningoencephalitis. In this review, we highlight key cryptococcal factors involved in various stages of cryptococcal-host interaction and pathogenesis.
Collapse
Affiliation(s)
| | | | - Tamara L. Doering
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA; (E.A.G.); (H.L.C.)
| |
Collapse
|
6
|
PDK1/mTOR Signaling in Myeloid Cells Differentially Regulates the Early and Late Stages of Sepsis. Mediators Inflamm 2020; 2020:5437175. [PMID: 32774145 PMCID: PMC7397376 DOI: 10.1155/2020/5437175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/08/2020] [Accepted: 07/06/2020] [Indexed: 11/20/2022] Open
Abstract
The cecal ligation and perforation (CLP) model is the gold standard for the polymicrobial sepsis. In the CLP mice, the myeloid cells play an important role in septic shock. The phenotypes and the activation state of the macrophage and neutrophil correlate with their metabolism. In the present study, we generated the specific myeloid deletion of PDK1 and mTOR mice, which was the important regulator of metabolic signaling. We found that the deletion of PDK1 in the myeloid cells could aggravate the early septic shock in the CLP mice, as well as the deletion of mTORC1 and mTORC2. Moreover, PDK1 deletion attenuated the inflammation induced by LPS in the late stage on CLP mice, which was exacerbated in mTORC1 and mTORC2 knockout mice. Both PDK1 and mTORC1/2 could not only regulate the cellular metabolism but also play important roles on the myeloid cells in the secondary stimulation of sepsis. The present study will provide a theoretical prospect for the therapy of the septic shock in different stages.
Collapse
|
7
|
Howard KC, Dennis EK, Watt DS, Garneau-Tsodikova S. A comprehensive overview of the medicinal chemistry of antifungal drugs: perspectives and promise. Chem Soc Rev 2020; 49:2426-2480. [PMID: 32140691 DOI: 10.1039/c9cs00556k] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The emergence of new fungal pathogens makes the development of new antifungal drugs a medical imperative that in recent years motivates the talents of numerous investigators across the world. Understanding not only the structural families of these drugs but also their biological targets provides a rational means for evaluating the merits and selectivity of new agents for fungal pathogens and normal cells. An equally important aspect of modern antifungal drug development takes a balanced look at the problems of drug potency and drug resistance. The future development of new antifungal agents will rest with those who employ synthetic and semisynthetic methodology as well as natural product isolation to tackle these problems and with those who possess a clear understanding of fungal cell architecture and drug resistance mechanisms. This review endeavors to provide an introduction to a growing and increasingly important literature, including coverage of the new developments in medicinal chemistry since 2015, and also endeavors to spark the curiosity of investigators who might enter this fascinatingly complex fungal landscape.
Collapse
Affiliation(s)
- Kaitlind C Howard
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596, USA.
| | | | | | | |
Collapse
|
8
|
Rauseo AM, Coler-Reilly A, Larson L, Spec A. Hope on the Horizon: Novel Fungal Treatments in Development. Open Forum Infect Dis 2020; 7:ofaa016. [PMID: 32099843 PMCID: PMC7031074 DOI: 10.1093/ofid/ofaa016] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 01/09/2020] [Indexed: 12/19/2022] Open
Abstract
The treatment of invasive fungal infections remains challenging due to limitations in currently available antifungal therapies including toxicity, interactions, restricted routes of administration, and drug resistance. This review focuses on novel therapies in clinical development, including drugs and a device. These drugs have novel mechanisms of action to overcome resistance, and some offer new formulations providing distinct advantages over current therapies to improve safety profiles and reduce interactions. Among agents that target the cell wall, 2 glucan synthesis inhibitors are discussed (rezafungin and ibrexafungerp), as well as fosmanogepix and nikkomycin Z. Agents that target the cell membrane include 3 fourth-generation azoles, oral encochleated amphotericin B, and aureobasidin A. Among agents with intracellular targets, we will review olorofim, VL-2397, T-2307, AR-12, and MGCD290. In addition, we will describe neurapheresis, a device used as adjunctive therapy for cryptococcosis. With a field full of novel treatments for fungal infections, the future looks promising.
Collapse
Affiliation(s)
- Adriana M Rauseo
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Lindsey Larson
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Andrej Spec
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
9
|
Maliehe M, Ntoi MA, Lahiri S, Folorunso OS, Ogundeji AO, Pohl CH, Sebolai OM. Environmental Factors That Contribute to the Maintenance of Cryptococcus neoformans Pathogenesis. Microorganisms 2020; 8:microorganisms8020180. [PMID: 32012843 PMCID: PMC7074686 DOI: 10.3390/microorganisms8020180] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/04/2019] [Accepted: 12/11/2019] [Indexed: 02/07/2023] Open
Abstract
The ability of microorganisms to colonise and display an intracellular lifestyle within a host body increases their fitness to survive and avoid extinction. This host–pathogen association drives microbial evolution, as such organisms are under selective pressure and can become more pathogenic. Some of these microorganisms can quickly spread through the environment via transmission. The non-transmittable fungal pathogens, such as Cryptococcus, probably return into the environment upon decomposition of the infected host. This review analyses whether re-entry of the pathogen into the environment causes restoration of its non-pathogenic state or whether environmental factors and parameters assist them in maintaining pathogenesis. Cryptococcus (C.) neoformans is therefore used as a model organism to evaluate the impact of environmental stress factors that aid the survival and pathogenesis of C. neoformans intracellularly and extracellularly.
Collapse
|
10
|
Abstract
Melanins are dark green, brown, or black pigments that serve as antioxidant, reactive oxygen species (ROS) scavengers that protect fungal pathogens from radiation and host immune responses. Cryptococcus neoformans, the major etiological agent of fungal meningoencephalitis, also utilizes melanin as a key virulence factor. In this basidiomycete pathogen, melanin production is regulated by the cAMP and high-osmolarity glycerol response (HOG) pathways, and yet its complex signaling networks remain poorly described. In this study, we uncovered novel melanin synthesis regulatory networks consisting of core transcription factors (TFs), including Bzp4, Usv101, Hob1, and Mbs1, and core kinases Gsk3 and Kic1. These networks were identified through coupling systematic analyses of the expression and epistatic relationships of TF and kinase mutant libraries in the presence of diverse melanin substrates with transcriptome profiling of the core TF mutants. Thus, this report provides comprehensive insight into the melanin-regulating pathways in C. neoformans and other fungal pathogens. Melanin is an antioxidant polyphenol pigment required for the pathogenicity of many fungal pathogens, but comprehensive regulatory mechanisms remain unidentified. In this study, we systematically analyzed melanin-regulating signaling pathways in Cryptococcus neoformans and identified four melanin-regulating core transcription factors (TFs), Bzp4, Usv101, Mbs1, and Hob1, required for induction of the laccase gene (LAC1). Bzp4, Usv101, and Mbs1 independently regulate LAC1 induction, whereas Hob1 controls Bzp4 and Usv101 expression. Both Bzp4 and Usv101 are localized in the cytoplasm under nutrient-rich conditions (i.e., in the presence of yeast extract-peptone-dextrose [YPD] medium) but translocate into the nucleus upon nutrient starvation (i.e., in the presence of yeast nitrogen base [YNB] medium without glucose), and Mbs1 is constitutively localized in the nucleus. Notably, the cAMP pathway is not involved in regulation of the four TFs, but the high-osmolarity glycerol response (HOG) pathway negatively regulates induction of BZP4 and LAC1. Next, we searched for potential kinases upstream of the core TFs and identified nine core kinases; their deletion led to defective melanin production and LAC1 induction. Deletion of GSK3 or KIC1 abolished induction of LAC1 and BZP4 and perturbed nuclear translocation of Bzp4. Notably, Gsk3 also regulated expression of HOB1, USV101, and MBS1, indicating that it is a critical melanin-regulating kinase. Finally, an RNA sequencing-based transcriptome analysis of the wild-type strain and of bzp4Δ, usv101Δ, hob1Δ, and mbs1Δ strains under nutrient-rich and nutrient-starved conditions revealed that the melanin-regulating core TFs govern redundant and distinct classes of genes involved in a variety of biological processes.
Collapse
|
11
|
Leipheimer J, Bloom ALM, Panepinto JC. Protein Kinases at the Intersection of Translation and Virulence. Front Cell Infect Microbiol 2019; 9:318. [PMID: 31572689 PMCID: PMC6749009 DOI: 10.3389/fcimb.2019.00318] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 08/26/2019] [Indexed: 12/14/2022] Open
Abstract
As free living organisms, fungi are challenged with a variety of environmental insults that threaten their cellular processes. In some cases, these challenges mimic conditions present within mammals, resulting in the accidental selection of virulence factors over evolutionary time. Be it within a host or the soil, fungi must contend with environmental challenges through the production of stress effector proteins while maintaining factors required for viability in any condition. Initiation and upkeep of this balancing act is mainly under the control of kinases that affect the propensity and selectivity of protein translation. This review will focus on kinases in pathogenic fungi that facilitate a virulence phenotype through translational control.
Collapse
Affiliation(s)
- Jay Leipheimer
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Amanda L M Bloom
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - John C Panepinto
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
12
|
Matsumoto Y, Ishii M, Shimizu K, Kawamoto S, Sekimizu K. [A Silkworm Infection Model to Evaluate Antifungal Drugs for Cryptococcosis]. Med Mycol J 2018; 58:E131-E137. [PMID: 29187715 DOI: 10.3314/mmj.17.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The development of effective drugs against fungal diseases involves performing infection experiments in animals to evaluate candidate therapeutic compounds. Cryptococcus neoformans is a pathogenic fungus that causes deep mycosis, resulting in respiratory illness and meningitis. Here we describe a silkworm system established to evaluate the safety and efficacy of therapeutic drugs against infection by Cryptococcus neoformans and the advantages of this system over other animal models. The silkworm assay system has two major advantages: 1) silkworms are less expensive to rear and their use is less problematic than that of mammals in terms of animal welfare, and 2) in vivo screenings for identifying candidate drugs can be easily performed using a large number of silkworms. The pharmacokinetics of compounds are consistent between silkworms and mammals. Moreover, the ED50 values of antibiotics are concordant between mammalian and silkworm infection models. Furthermore, the body size of silkworms makes them easy to handle in experimental procedures compared with other invertebrate infectious experimental systems, and accurate amounts of pathogens and chemicals can be injected fairly easily. These advantages of silkworms as a host animal make them useful for screening candidate drugs for cryptococcosis.
Collapse
Affiliation(s)
| | - Masaki Ishii
- Teikyo University Institute of Medical Mycology.,Genome Pharmaceuticals Institute Co. Ltd
| | - Kiminori Shimizu
- Department of Biological Science and Technology, Tokyo University of Science
| | | | - Kazuhisa Sekimizu
- Teikyo University Institute of Medical Mycology.,Genome Pharmaceuticals Institute Co. Ltd
| |
Collapse
|
13
|
Mismatch Repair of DNA Replication Errors Contributes to Microevolution in the Pathogenic Fungus Cryptococcus neoformans. mBio 2017; 8:mBio.00595-17. [PMID: 28559486 PMCID: PMC5449657 DOI: 10.1128/mbio.00595-17] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
The ability to adapt to a changing environment provides a selective advantage to microorganisms. In the case of many pathogens, a large change in their environment occurs when they move from a natural setting to a setting within a human host and then during the course of disease development to various locations within that host. Two clinical isolates of the human fungal pathogen Cryptococcus neoformans were identified from a collection of environmental and clinical strains that exhibited a mutator phenotype, which is a phenotype which provides the ability to change rapidly due to the accumulation of DNA mutations at high frequency. Whole-genome analysis of these strains revealed mutations in MSH2 of the mismatch repair pathway, and complementation confirmed that these mutations are responsible for the mutator phenotype. Comparison of mutation frequencies in deletion strains of eight mismatch repair pathway genes in C. neoformans showed that the loss of three of them, MSH2, MLH1, and PMS1, results in an increase in mutation rates. Increased mutation rates enable rapid microevolution to occur in these strains, generating phenotypic variations in traits associated with the ability to grow in vivo, in addition to allowing rapid generation of resistance to antifungal agents. Mutation of PMS1 reduced virulence, whereas mutation of MSH2 or MLH1 had no effect on the level of virulence. These findings thus support the hypothesis that this pathogenic fungus can take advantage of a mutator phenotype in order to cause disease but that it can do so only in specific pathways that lead to a mutator trait without a significant tradeoff in fitness. Fungi account for a large number of infections that are extremely difficult to treat; superficial fungal infections affect approximately 1.7 billion (25%) of the general population worldwide, and systemic fungal diseases result in an unacceptably high mortality rate. How fungi adapt to their hosts is not fully understood. This research investigated the role of changes to DNA sequences in adaption to the host environment and the ability to cause disease in Cryptococcus neoformans, one of the world’s most common and most deadly fungal pathogens. The study results showed that microevolutionary rates are enhanced in either clinical isolates or in gene deletion strains with msh2 mutations. This gene has similar functions in regulating the rapid emergence of antifungal drug resistance in a distant fungal relative of C. neoformans, the pathogen Candida glabrata. Thus, microevolution resulting from enhanced mutation rates may be a common contributor to fungal pathogenesis.
Collapse
|
14
|
Bloom ALM, Leipheimer J, Panepinto JC. mRNA decay: an adaptation tool for the environmental fungal pathogen Cryptococcus neoformans. WILEY INTERDISCIPLINARY REVIEWS-RNA 2017; 8. [PMID: 28524625 DOI: 10.1002/wrna.1424] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/05/2017] [Accepted: 04/06/2017] [Indexed: 12/22/2022]
Abstract
Fungi are ubiquitous in the environment and humans constantly encounter them in the soil, air, water, and food. The vast majority of these interactions are inconsequential. However, in the context of immunodeficiency precipitated by HIV infection, hematologic malignancy, or transplantation, a small subset of fungi can cause devastating, systemic infection. The most deadly of the opportunistic environmental fungi, Cryptococcus neoformans, is estimated to cause hundreds of thousands of deaths per year, mostly in the context of HIV co-infection. The cellular processes that mediate adaptation to the host environment are of great interest as potential novel therapeutic targets. One such cellular process important for host adaptation is mRNA decay, which mediates the specific degradation of subsets of functionally related mRNAs in response to stressors relevant to pathogenesis, including human core body temperature, carbon limitation, and reactive oxygen stress. Thus, for C. neoformans, host adaptation requires mRNA decay to mediate rapid transcriptome remodeling in the face of stressors encountered in the host. Several nodes of stress-responsive signaling that govern the stress-responsive transcriptome also control the decay rate of mRNAs cleared from the ribosome during stress, suggesting an additional layer of coupling between mRNA synthesis and decay that allows C. neoformans to be a successful pathogen of humans. WIREs RNA 2017, 8:e1424. doi: 10.1002/wrna.1424 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Amanda L M Bloom
- Witebsky Center for Microbial Pathogenesis and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Jay Leipheimer
- Witebsky Center for Microbial Pathogenesis and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - John C Panepinto
- Witebsky Center for Microbial Pathogenesis and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| |
Collapse
|
15
|
The Celecoxib Derivative AR-12 Has Broad-Spectrum Antifungal Activity In Vitro and Improves the Activity of Fluconazole in a Murine Model of Cryptococcosis. Antimicrob Agents Chemother 2016; 60:7115-7127. [PMID: 27645246 DOI: 10.1128/aac.01061-16] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/04/2016] [Indexed: 12/24/2022] Open
Abstract
Only one new class of antifungal drugs has been introduced into clinical practice in the last 30 years, and thus the identification of small molecules with novel mechanisms of action is an important goal of current anti-infective research. Here, we describe the characterization of the spectrum of in vitro activity and in vivo activity of AR-12, a celecoxib derivative which has been tested in a phase I clinical trial as an anticancer agent. AR-12 inhibits fungal acetyl coenzyme A (acetyl-CoA) synthetase in vitro and is fungicidal at concentrations similar to those achieved in human plasma. AR-12 has a broad spectrum of activity, including activity against yeasts (e.g., Candida albicans, non-albicans Candida spp., Cryptococcus neoformans), molds (e.g., Fusarium, Mucor), and dimorphic fungi (Blastomyces, Histoplasma, and Coccidioides) with MICs of 2 to 4 μg/ml. AR-12 is also active against azole- and echinocandin-resistant Candida isolates, and subinhibitory AR-12 concentrations increase the susceptibility of fluconazole- and echinocandin-resistant Candida isolates. Finally, AR-12 also increases the activity of fluconazole in a murine model of cryptococcosis. Taken together, these data indicate that AR-12 represents a promising class of small molecules with broad-spectrum antifungal activity.
Collapse
|
16
|
Banerjee D, Bloom ALM, Panepinto JC. Opposing PKA and Hog1 signals control the post-transcriptional response to glucose availability in Cryptococcus neoformans. Mol Microbiol 2016; 102:306-320. [PMID: 27387858 DOI: 10.1111/mmi.13461] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2016] [Indexed: 12/17/2022]
Abstract
The pathogenic fungus Cryptococcus neoformans must adapt to glucose-limited conditions in the lung and glucose replete conditions upon dissemination to the brain. We report that glucose controls ribosome biogenesis and translation by modulating mRNA decay through a balance of PKA and Hog1 signalling. Glucose signalling through PKA stabilized ribosomal protein (RP) mRNAs whereas glucose starvation destabilized RP transcripts through Hog1. Glucose starvation-induced oxidative stress response genes, and treatment of glucose-fed cells with reactive oxygen species (ROS) generating compounds repressed RP transcripts, both of which were dependent on Hog1. Stabilization of RP transcripts led to retention of polysomes in a hog1Δ mutant, whereas stabilization of RP transcripts by cyclic AMP did not affect translation repression, suggesting that Hog1 alone signals translation repression. In sum, this work describes a novel antagonism between PKA and Hog1 controlling ribosome biogenesis via mRNA stability in response to glucose availability in this important human pathogen.
Collapse
Affiliation(s)
- Dithi Banerjee
- Department of Microbiology and Immunology, Witebsky Center for Microbial Pathogenesis and Immunology, State University of New York at Buffalo, Buffalo, NY, 14214, USA
| | - Amanda L M Bloom
- Department of Microbiology and Immunology, Witebsky Center for Microbial Pathogenesis and Immunology, State University of New York at Buffalo, Buffalo, NY, 14214, USA
| | - John C Panepinto
- Department of Microbiology and Immunology, Witebsky Center for Microbial Pathogenesis and Immunology, State University of New York at Buffalo, Buffalo, NY, 14214, USA.
| |
Collapse
|
17
|
Koselny K, Green J, Favazzo L, Glazier V, DiDone L, Ransford S, Krysan DJ. Antitumor/Antifungal Celecoxib Derivative AR-12 is a Non-Nucleoside Inhibitor of the ANL-Family Adenylating Enzyme Acetyl CoA Synthetase. ACS Infect Dis 2016; 2:268-280. [PMID: 27088128 PMCID: PMC4828684 DOI: 10.1021/acsinfecdis.5b00134] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Indexed: 02/08/2023]
Abstract
AR-12/OSU-03012 is an antitumor celecoxib-derivative that has progressed to Phase I clinical trial as an anticancer agent and has activity against a number of infectious agents including fungi, bacteria and viruses. However, the mechanism of these activities has remained unclear. Based on a chemical-genetic profiling approach in yeast, we have found that AR-12 is an ATP-competitive, time-dependent inhibitor of yeast acetyl coenzyme A synthetase. AR-12-treated fungal cells show phenotypes consistent with the genetic reduction of acetyl CoA synthetase activity, including induction of autophagy, decreased histone acetylation, and loss of cellular integrity. In addition, AR-12 is a weak inhibitor of human acetyl CoA synthetase ACCS2. Acetyl CoA synthetase activity is essential in many fungi and parasites. In contrast, acetyl CoA is primarily synthesized by an alternate enzyme, ATP-citrate lyase, in mammalian cells. Taken together, our results indicate that AR-12 is a non-nucleoside acetyl CoA synthetase inhibitor and that acetyl CoA synthetase may be a feasible antifungal drug target.
Collapse
Affiliation(s)
- Kristy Koselny
- Department of Pediatrics and Department of
Microbiology/Immunology, University of Rochester
School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York 14642, United
States
| | - Julianne Green
- Department of Pediatrics and Department of
Microbiology/Immunology, University of Rochester
School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York 14642, United
States
| | - Lacey Favazzo
- Department of Pediatrics and Department of
Microbiology/Immunology, University of Rochester
School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York 14642, United
States
| | - Virginia
E. Glazier
- Department of Pediatrics and Department of
Microbiology/Immunology, University of Rochester
School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York 14642, United
States
| | - Louis DiDone
- Department of Pediatrics and Department of
Microbiology/Immunology, University of Rochester
School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York 14642, United
States
| | - Shea Ransford
- Department of Pediatrics and Department of
Microbiology/Immunology, University of Rochester
School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York 14642, United
States
| | - Damian J. Krysan
- Department of Pediatrics and Department of
Microbiology/Immunology, University of Rochester
School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, New York 14642, United
States
| |
Collapse
|
18
|
Needle-Free Delivery of Acetalated Dextran-Encapsulated AR-12 Protects Mice from Francisella tularensis Lethal Challenge. Antimicrob Agents Chemother 2016; 60:2052-62. [PMID: 26787696 DOI: 10.1128/aac.02228-15] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 01/06/2016] [Indexed: 11/20/2022] Open
Abstract
Francisella tularensiscauses tularemia and is a potential biothreat. Given the limited antibiotics for treating tularemia and the possible use of antibiotic-resistant strains as a biowarfare agent, new antibacterial agents are needed. AR-12 is an FDA-approved investigational new drug (IND) compound that induces autophagy and has shown host-directed, broad-spectrum activityin vitroagainstSalmonella entericaserovar Typhimurium andF. tularensis We have shown that AR-12 encapsulated within acetalated dextran (Ace-DEX) microparticles (AR-12/MPs) significantly reduces host cell cytotoxicity compared to that with free AR-12, while retaining the ability to controlS.Typhimurium within infected human macrophages. In the present study, the toxicity and efficacy of AR-12/MPs in controlling virulent type AF. tularensisSchuS4 infection were examinedin vitroandin vivo No significant toxicity of blank MPs or AR-12/MPs was observed in lung histology sections when the formulations were given intranasally to uninfected mice. In histology sections from the lungs of intranasally infected mice treated with the formulations, increased macrophage infiltration was observed for AR-12/MPs, with or without suboptimal gentamicin treatment, but not for blank MPs, soluble AR-12, or suboptimal gentamicin alone. AR-12/MPs dramatically reduced the burden ofF. tularensisin infected human macrophages, in a manner similar to that of free AR-12. However,in vivo, AR-12/MPs significantly enhanced the survival ofF. tularensisSchuS4-infected mice compared to that seen with free AR-12. In combination with suboptimal gentamicin treatment, AR-12/MPs further improved the survival ofF. tularensisSchuS4-infected mice. These studies provide support for Ace-DEX-encapsulated AR-12 as a promising new therapeutic agent for tularemia.
Collapse
|
19
|
Ishii M, Matsumoto Y, Sekimizu K. Usefulness of silkworm as a host animal for understanding pathogenicity of Cryptococcus neoformans. Drug Discov Ther 2016; 10:9-13. [PMID: 26902902 DOI: 10.5582/ddt.2016.01015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
We propose Cryptococcus neoformans infection model using silkworm for understanding cryptococcosis and screening of therapeutically effective antibiotics. Silkworm is an insect whose rearing methods were established through a long history of the sericulture industry. Silkworm facilitates experiments using a large number of individuals because of low cost for rearing and few ethical problems caused by killing animals. Silkworm can be reared at 37˚C to perform infection experiments at same temperature to human body. Injection of accurate amounts of samples into hemolymph of silkworm by usual syringes is easy to be done since silkworm has an appropriate size to handle. Moreover two injection methods, injection into hemolymph and intestine, are distinguishable for silkworms. The former is correspondent to intravenous injection, and the latter is to oral administration in humans. Taking these advantages of silkworms as host animals, it is possible to evaluate the virulence factors in C. neoformans and the therapeutic efficacy of antifungal agents.
Collapse
Affiliation(s)
- Masaki Ishii
- Laboratory of Microbiology, Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | | | | |
Collapse
|
20
|
Hartland K, Pu J, Palmer M, Dandapani S, Moquist PN, Munoz B, DiDone L, Schreiber SL, Krysan DJ. High-Throughput Screen in Cryptococcus neoformans Identifies a Novel Molecular Scaffold That Inhibits Cell Wall Integrity Pathway Signaling. ACS Infect Dis 2016; 2:93-102. [PMID: 26807437 PMCID: PMC4709821 DOI: 10.1021/acsinfecdis.5b00111] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Indexed: 02/07/2023]
Abstract
![]()
Cryptococcus neoformans is one of the most important
human fungal pathogens; however, no new therapies have been developed
in over 50 years. Fungicidal activity is crucially important for an
effective anticryptococal agent and, therefore, we screened 361,675
molecules against C. neoformans using an adenylate
kinase release assay that specifically detects fungicidal activity.
A set of secondary assays narrowed the set of hits to molecules that
interfere with fungal cell wall integrity and identified three benzothioureas
with low in vitro mammalian toxicity and good in vitro anticryptococcal
(minimum inhibitory concentration = 4 μg/mL). This scaffold
inhibits signaling through the cell wall integrity MAP kinase cascade.
Structure–activity studies indicate that the thiocarbonyl moiety
is crucial for activity. Genetic and biochemical data suggest that
benzothioureas inhibit signaling upstream of the kinase cascade. Thus,
the benzothioureas appear to be a promising new scaffold for further
exploration in the search for new anticryptococcal agents.
Collapse
Affiliation(s)
- Kate Hartland
- Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, 7 Cambridge
Center, Cambridge, Massachusetts 02142, United States
| | - Jun Pu
- Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, 7 Cambridge
Center, Cambridge, Massachusetts 02142, United States
| | - Michelle Palmer
- Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, 7 Cambridge
Center, Cambridge, Massachusetts 02142, United States
| | - Sivaraman Dandapani
- Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, 7 Cambridge
Center, Cambridge, Massachusetts 02142, United States
| | - Philip N. Moquist
- Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, 7 Cambridge
Center, Cambridge, Massachusetts 02142, United States
| | - Benito Munoz
- Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, 7 Cambridge
Center, Cambridge, Massachusetts 02142, United States
| | | | - Stuart L. Schreiber
- Center for the Science of Therapeutics, Broad Institute of Harvard and MIT, 7 Cambridge
Center, Cambridge, Massachusetts 02142, United States
| | | |
Collapse
|
21
|
Collier MA, Peine KJ, Gautam S, Oghumu S, Varikuti S, Borteh H, Papenfuss TL, Sataoskar AR, Bachelder EM, Ainslie KM. Host-mediated Leishmania donovani treatment using AR-12 encapsulated in acetalated dextran microparticles. Int J Pharm 2016; 499:186-194. [PMID: 26768723 DOI: 10.1016/j.ijpharm.2016.01.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 12/29/2015] [Accepted: 01/04/2016] [Indexed: 12/15/2022]
Abstract
Leishmaniasis is a disease caused by parasites of Leishmania sp., which effects nearly 12 million people worldwide and is associated with treatment complications due to widespread parasite resistance toward pathogen-directed therapeutics. The current treatments for visceral leishmaniasis (VL), the systemic form of the disease, involve pathogen-mediated drugs and have long treatment regimens, increasing the risk of forming resistant strains. One way to limit emergence of resistant pathogens is through the use of host-mediated therapeutics. The host-mediated therapeutic AR-12, which is FDA IND-approved for cancer treatment, has shown activity against a broad spectrum of intracellular pathogens; however, due to hydrophobicity and toxicity, it is difficult to reach therapeutic doses. We have formulated AR-12 into microparticles (AR-12/MPs) using the novel biodegradable polymer acetalated dextran (Ace-DEX) and used this formulation for the systemic treatment of VL. Treatment with AR-12/MPs significantly reduced liver, spleen, and bone marrow parasite loads in infected mice, while combinatorial therapies with amphotericin B had an even more significant effect. Overall, AR-12/MPs offer a unique, host-mediated therapy that could significantly reduce the emergence of drug resistance in the treatment of VL.
Collapse
Affiliation(s)
- M A Collier
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - K J Peine
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - S Gautam
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - S Oghumu
- Department of Pathology, The Ohio State's Wexner Medical Center, The Ohio State University, Columbus, OH 43210, United States
| | - S Varikuti
- Department of Pathology, The Ohio State's Wexner Medical Center, The Ohio State University, Columbus, OH 43210, United States
| | - H Borteh
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - T L Papenfuss
- Department of Pathology, The Ohio State's Wexner Medical Center, The Ohio State University, Columbus, OH 43210, United States
| | - A R Sataoskar
- College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, United States
| | - E M Bachelder
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - K M Ainslie
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States.
| |
Collapse
|
22
|
Turrà D, Segorbe D, Di Pietro A. Protein kinases in plant-pathogenic fungi: conserved regulators of infection. ANNUAL REVIEW OF PHYTOPATHOLOGY 2014; 52:267-88. [PMID: 25090477 DOI: 10.1146/annurev-phyto-102313-050143] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Phytopathogenic fungi have evolved an amazing diversity of infection modes and nutritional strategies, yet the signaling pathways that govern pathogenicity are remarkably conserved. Protein kinases (PKs) catalyze the reversible phosphorylation of proteins, regulating a variety of cellular processes. Here, we present an overview of our current understanding of the different classes of PKs that contribute to fungal pathogenicity on plants and of the mechanisms that regulate and coordinate PK activity during infection-related development. In addition to the well-studied PK modules, such as MAPK (mitogen-activated protein kinase) and cAMP (cyclic adenosine monophosphate)-PKA (protein kinase A) cascades, we also discuss new PK pathways that have emerged in recent years as key players of pathogenic development and disease. Understanding how conserved PK signaling networks have been recruited during the evolution of fungal pathogenicity not only advances our knowledge of the highly elaborate infection process but may also lead to the development of novel strategies for the control of plant disease.
Collapse
Affiliation(s)
- David Turrà
- Departamento de Genética and Campus de Excelencia Agroalimentario (ceiA3), Universidad de Córdoba, 14071 Córdoba, Spain; , ,
| | | | | |
Collapse
|
23
|
Abstract
Cryptococcus species are fungal pathogens that are a leading cause of mortality. Initial inoculation is through the pulmonary route and, if disseminated, results in severe invasive infection including meningoencephalitis. Macrophages are the dominant phagocytic cell that interacts with Cryptococcus. Emerging theories suggest that Cryptococcus microevolution in macrophages is linked to survival and virulence within the host. In addition, Cryptococcus elaborates virulence factors as well as usurps host machinery to establish macrophage activation states that are permissive to intracellular survival and replication. In this review, we provide an update of the recent findings pertaining to macrophage interaction with Cryptococcus and focus on new avenues for biomedical research.
Collapse
|
24
|
Pastor-Flores D, Schulze JO, Bahí A, Giacometti R, Ferrer-Dalmau J, Passeron S, Engel M, Süß E, Casamayor A, Biondi RM. PIF-pocket as a target for C. albicans Pkh selective inhibitors. ACS Chem Biol 2013; 8:2283-92. [PMID: 23911092 DOI: 10.1021/cb400452z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The phosphoinositide-dependent protein kinase 1, PDK1, is a master kinase that phosphorylates the activation loop of up to 23 AGC kinases. S. cerevisiae has three PDK1 orthologues, Pkh1-3, which also phosphorylate AGC kinases (e.g., Ypk, Tpk, Pkc1, and Sch9). Pkh1 and 2 are redundant proteins involved in multiple essential cellular functions, including endocytosis and cell wall integrity. Based on similarities with the budding yeast, the Pkh of fungal infectious species was postulated as a novel target for antifungals. Here, we found that depletion of Pkh eventually induces oxidative stress and DNA double-strand breaks, leading to programmed cell death. This finding supports Pkh as an antifungal target since pharmacological inhibition of Pkh would lead to the death of yeast cells, the ultimate goal of antifungals. It was therefore of interest to further investigate the possibility to develop Pkh inhibitors with selectivity for Candida Pkh that would not inhibit the human ortholog. Here, we describe C. albicans Pkh2 biochemically, structurally and by using chemical probes in comparison to human PDK1. We found that a regulatory site on the C. albicans Pkh2 catalytic domain, the PIF-pocket, diverges from human PDK1. Indeed, we identified and characterized PS77, a new small allosteric inhibitor directed to the PIF-pocket, which has increased selectivity for C. albicans Pkh2. Together, our results describe novel features of the biology of Pkh and chemical biology approaches that support the validation of Pkh as a drug target for selective antifungals.
Collapse
Affiliation(s)
- Daniel Pastor-Flores
- Research Group PhosphoSites,
Medizinische Klinik 1, Universitätsklinikum Frankfurt, Theodor-Stern-Kai
7, 60590 Frankfurt, Germany
| | - Jörg O. Schulze
- Research Group PhosphoSites,
Medizinische Klinik 1, Universitätsklinikum Frankfurt, Theodor-Stern-Kai
7, 60590 Frankfurt, Germany
| | - Anna Bahí
- Departament de Bioquímica
i Biologia Molecular, Facultat de Veterinària, Universitat
Autònoma de Barcelona, Cerdanyola 08193, Barcelona, Spain
- Institut de Biotecnologia i Biomedicina,
Universitat Autònoma de Barcelona, Cerdanyola 08193, Barcelona,
Spain
| | - Romina Giacometti
- Cátedra de
Bioquímica,
Facultad de Agronomía, Universidad de Buenos Aires, C1417DSE
Buenos Aires, Argentina
| | - Jofre Ferrer-Dalmau
- Departament de Bioquímica
i Biologia Molecular, Facultat de Veterinària, Universitat
Autònoma de Barcelona, Cerdanyola 08193, Barcelona, Spain
- Institut de Biotecnologia i Biomedicina,
Universitat Autònoma de Barcelona, Cerdanyola 08193, Barcelona,
Spain
| | - Susana Passeron
- Cátedra de
Bioquímica,
Facultad de Agronomía, Universidad de Buenos Aires, C1417DSE
Buenos Aires, Argentina
| | - Matthias Engel
- Pharmaceutical and Medicinal
Chemistry, Saarland University, P.O. Box 151150, D-66041 Saarbrücken,
Germany
| | - Evelyn Süß
- Research Group PhosphoSites,
Medizinische Klinik 1, Universitätsklinikum Frankfurt, Theodor-Stern-Kai
7, 60590 Frankfurt, Germany
| | - Antonio Casamayor
- Departament de Bioquímica
i Biologia Molecular, Facultat de Veterinària, Universitat
Autònoma de Barcelona, Cerdanyola 08193, Barcelona, Spain
- Institut de Biotecnologia i Biomedicina,
Universitat Autònoma de Barcelona, Cerdanyola 08193, Barcelona,
Spain
| | - Ricardo M. Biondi
- Research Group PhosphoSites,
Medizinische Klinik 1, Universitätsklinikum Frankfurt, Theodor-Stern-Kai
7, 60590 Frankfurt, Germany
| |
Collapse
|
25
|
Stress signaling pathways for the pathogenicity of Cryptococcus. EUKARYOTIC CELL 2013; 12:1564-77. [PMID: 24078305 DOI: 10.1128/ec.00218-13] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Sensing, responding, and adapting to the surrounding environment are crucial for all living organisms to survive, proliferate, and differentiate in their biological niches. This ability is also essential for Cryptococcus neoformans and its sibling species Cryptococcus gattii, as these pathogens have saprobic and parasitic life cycles in natural and animal host environments. The ability of Cryptococcus to cause fatal meningoencephalitis is highly related to its capability to remodel and optimize its metabolic and physiological status according to external cues. These cues act through multiple stress signaling pathways through a panoply of signaling components, including receptors/sensors, small GTPases, secondary messengers, kinases, transcription factors, and other miscellaneous adaptors or regulators. In this minireview, we summarize and highlight the importance of several stress signaling pathways that influence the pathogenicity of Cryptococcus and discuss future challenges in these areas.
Collapse
|
26
|
Bloom ALM, Solomons JTG, Havel VE, Panepinto JC. Uncoupling of mRNA synthesis and degradation impairs adaptation to host temperature in Cryptococcus neoformans. Mol Microbiol 2013; 89:65-83. [PMID: 23659661 DOI: 10.1111/mmi.12258] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2013] [Indexed: 11/25/2022]
Abstract
The pathogenic fungus Cryptococcus neoformans must overcome multiple stressors to cause disease in its human host. In this study, we report that C. neoformans rapidly and transiently repressed ribosomal protein (RP) transcripts during a transition from 30°C to host temperature. This repression was accompanied by accelerated mRNA degradation mediated by the major deadenylase, Ccr4, and influenced by the dissociable RNA polymerase II subunit, Rpb4. Destabilization and deadenylation of RP transcripts were impaired in an rpb4Δ mutant, suggesting that Rpb4 may be involved in host temperature-induced Ccr4-mediated decay. Accelerated decay of ER stress transcripts 1 h following a shift to host temperature was also impaired in the rpb4Δ mutant. In response to host temperature, Rpb4 moved from the nucleus to the cytoplasm, supporting a role for Rpb4 in coupling transcription and degradation. The PKH signalling pathway was implicated as a regulator of accelerated degradation of the RP transcripts, but not of the ER stress transcripts, revealing a further level of specificity. When transcription and degradation were uncoupled by deletion of Rpb4, growth at host temperature was impaired and virulence was attenuated. These data suggest that mRNA synthesis and decay are coupled in C. neoformans via Rpb4, and this tight coordination promotes host-temperature adaptation and pathogenicity.
Collapse
Affiliation(s)
- Amanda L M Bloom
- Department of Microbiology and Immunology, Witebsky Center for Microbial Pathogenesis and Immunology, University at Buffalo, the State University of New York, Buffalo, NY, USA
| | | | | | | |
Collapse
|
27
|
Butts A, DiDone L, Koselny K, Baxter BK, Chabrier-Rosello Y, Wellington M, Krysan DJ. A repurposing approach identifies off-patent drugs with fungicidal cryptococcal activity, a common structural chemotype, and pharmacological properties relevant to the treatment of cryptococcosis. EUKARYOTIC CELL 2013; 12:278-87. [PMID: 23243064 PMCID: PMC3571299 DOI: 10.1128/ec.00314-12] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 12/10/2012] [Indexed: 11/20/2022]
Abstract
New, more accessible therapies for cryptococcosis represent an unmet clinical need of global importance. We took a repurposing approach to identify previously developed drugs with fungicidal activity toward Cryptococcus neoformans, using a high-throughput screening assay designed to detect drugs that directly kill fungi. From a set of 1,120 off-patent medications and bioactive molecules, we identified 31 drugs/molecules with fungicidal activity, including 15 drugs for which direct antifungal activity had not previously been reported. A significant portion of the drugs are orally bioavailable and cross the blood-brain barrier, features key to the development of a widely applicable anticryptococcal agent. Structural analysis of this set revealed a common chemotype consisting of a hydrophobic moiety linked to a basic amine, features that are common to drugs that cross the blood-brain barrier and access the phagolysosome, two important niches of C. neoformans. Consistent with their fungicidal activity, the set contains eight drugs that are either additive or synergistic in combination with fluconazole. Importantly, we identified two drugs, amiodarone and thioridazine, with activity against intraphagocytic C. neoformans. Finally, the set of drugs is also enriched for molecules that inhibit calmodulin, and we have confirmed that seven drugs directly bind C. neoformans calmodulin, providing a molecular target that may contribute to the mechanism of antifungal activity. Taken together, these studies provide a foundation for the optimization of the antifungal properties of a set of pharmacologically attractive scaffolds for the development of novel anticryptococcal therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Damian J. Krysan
- Pediatrics
- Microbiology/Immunology, University of Rochester, Rochester, New York, USA
| |
Collapse
|