1
|
Milsap D, Okuno M, Kigozi E, Mugabi T, Faizo S, Bajer A, Gakuru J, Bahr NC. Induction Treatment for HIV-Associated Cryptococcal Meningitis: Where Have We Been and Where Are We Going? Microorganisms 2025; 13:847. [PMID: 40284683 PMCID: PMC12029529 DOI: 10.3390/microorganisms13040847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 03/24/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
Cryptococcal meningitis remains a leading cause of morbidity and mortality among individuals with HIV/AIDS, particularly in resource-limited settings. Treatment begins with induction therapy followed by consolidation and maintenance. Evidence related to induction therapy has evolved significantly over the past decade. Current treatment relies primarily on three antifungal agents: amphotericin B, flucytosine, and fluconazole, each with distinct mechanisms of action and limitations. The World Health Organization's 2022 guidelines for induction therapy recommend a single high dose of liposomal amphotericin B combined with 14 days of flucytosine and fluconazole. The 2010 IDSA guidelines for induction therapy recommend amphotericin B deoxycholate and flucytosine for two weeks. The U.S. CDC/NIH/IDSA/HIVMA joint guidelines and the ECCM/ISHAM/ASM joint guidelines list both options, but the recommendation varies by setting resources (e.g., resource-limited vs. other). The newer treatment approaches (single high-dose liposomal amphotericin B) that are supported by trials such as AMBITION-cryptococcal meningitis have limited adoption in high-resource settings, with recent studies showing that only 14% of North American infectious disease providers have utilized the regimen. Adjunctive medications, such as dexamethasone, tamoxifen, and sertraline, have proven ineffective or harmful in clinical trials. This review underscores the ongoing challenges in cryptococcal meningitis treatment and the need for continued research to improve patient outcomes, tracing the evolution from past monotherapy approaches to current combination strategies while exploring future directions.
Collapse
Affiliation(s)
- Dominique Milsap
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; (D.M.); (M.O.)
| | - Madison Okuno
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; (D.M.); (M.O.)
| | - Enos Kigozi
- Infectious Diseases Institute, Makerere University, Kampala, Uganda; (E.K.); (T.M.); (S.F.); (J.G.)
- Clinical Epidemiology Unit, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Timothy Mugabi
- Infectious Diseases Institute, Makerere University, Kampala, Uganda; (E.K.); (T.M.); (S.F.); (J.G.)
| | - Ssekindi Faizo
- Infectious Diseases Institute, Makerere University, Kampala, Uganda; (E.K.); (T.M.); (S.F.); (J.G.)
| | - Aleksandra Bajer
- School of Medicine, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Jane Gakuru
- Infectious Diseases Institute, Makerere University, Kampala, Uganda; (E.K.); (T.M.); (S.F.); (J.G.)
| | - Nathan C. Bahr
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, 420 Delaware Street, SE, MMC 250, Minneapolis, MN 55455, USA
| |
Collapse
|
2
|
Yan ZZ, Hu HW, Xiong C, Peleg AY, Chen QL, Sáez-Sandino T, Maestre F, Delgado-Baquerizo M, Singh BK. Environmental microbiome, human fungal pathogens, and antimicrobial resistance. Trends Microbiol 2025; 33:112-129. [PMID: 39304419 DOI: 10.1016/j.tim.2024.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 09/22/2024]
Abstract
Traditionally, antifungal resistance (AFR) has received much less attention compared with bacterial resistance to antibiotics. However, global changes, pandemics, and emerging new fungal infections have highlighted global health consequences of AFR. The recent report of the World Health Organisation (WHO) has identified fungal priority pathogens, and recognised AFR among the greatest global health threats. This is particularly important given the significant increase in fungal infections linked to climate change and pandemics. Environmental factors play critical roles in AFR and fungal infections, as many clinically relevant fungal pathogens and AFR originate from the environment (mainly soil). In addition, the environment serves as a potential rich source for the discovery of new antifungal agents, including mycoviruses and bacterial probiotics, which hold promise for effective therapies. In this article, we summarise the environmental pathways of AFR development and spread among high priority fungal pathogens, and propose potential mechanisms of AFR development and spread. We identify a research priority list to address key knowledge gaps in our understanding of environmental AFR. Further, we propose an integrated roadmap for predictive risk management of AFR that is critical for effective surveillance and forecasting of public health outcomes under current and future climatic conditions.
Collapse
Affiliation(s)
- Zhen-Zhen Yan
- Hawkesbury Institute for the Environment, Western Sydney University, Penrith, Australia
| | - Hang-Wei Hu
- School of Agriculture, Food, and Ecosystem Science, Faculty of Science, The University of Melbourne, Victoria, Australia
| | - Chao Xiong
- Hawkesbury Institute for the Environment, Western Sydney University, Penrith, Australia
| | - Anton Y Peleg
- Department of Infectious Disease, The Alfred Hospital and Central Clinical School, Monash University, Victoria, Australia; Department of Microbiology, Monash University, Melbourne, Australia; Centre to Impact Antimicrobial Resistance, Monash University, Melbourne, Australia
| | - Qing-Lin Chen
- Key Laboratory of Urban Environment and Health, Ningbo Observation and Research Station, Institute of Urban Environment, Chinese Academy of Science, Xiamen, China; University of Chinese Academy of Sciences, Beijing, China
| | - Tadeo Sáez-Sandino
- Hawkesbury Institute for the Environment, Western Sydney University, Penrith, Australia
| | - Fernando Maestre
- Environmental Sciences and Engineering, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Kingdom of Saudi Arabia
| | - Manuel Delgado-Baquerizo
- Laboratorio de Biodiversidad y Funcionamiento Ecosistémico, Instituto de Recursos Naturales y Agrobiología de Sevilla (IRNAS), CSIC, Sevilla, Spain
| | - Brajesh K Singh
- Hawkesbury Institute for the Environment, Western Sydney University, Penrith, Australia.
| |
Collapse
|
3
|
Ball B, Sukumaran A, Pladwig S, Kazi S, Chan N, Honeywell E, Modrakova M, Geddes-McAlister J. Proteome signatures reveal homeostatic and adaptive oxidative responses by a putative co-chaperone, Wos2, to influence fungal virulence determinants in cryptococcosis. Microbiol Spectr 2024; 12:e0015224. [PMID: 38953322 PMCID: PMC11302251 DOI: 10.1128/spectrum.00152-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/27/2024] [Indexed: 07/04/2024] Open
Abstract
The increasing prevalence of invasive fungal pathogens is dramatically changing the clinical landscape of infectious diseases, posing an imminent threat to public health. Specifically, Cryptococcus neoformans, the human opportunistic pathogen, expresses elaborate virulence mechanisms and is equipped with sophisticated adaptation strategies to survive in harsh host environments. This study extensively characterizes Wos2, an Hsp90 co-chaperone homolog, featuring bilateral functioning for both cryptococcal adaptation and the resulting virulence response. In this study, we evaluated the proteome and secretome signatures associated with wos2 deletion in enriched and infection-mimicking conditions to reveal Wos2-dependent regulation of the oxidative stress response through global translational reprogramming. The wos2Δ strain demonstrates defective intracellular and extracellular antioxidant protection systems, measurable through a decreased abundance of critical antioxidant enzymes and reduced growth in the presence of peroxide stress. Additional Wos2-associated stress phenotypes were observed upon fungal challenge with heat shock, osmotic stress, and cell membrane stressors. We demonstrate the importance of Wos2 for intracellular lifestyle of C. neoformans during in vitro macrophage infection and provide evidence for reduced phagosomal replication levels associated with wos2Δ. Accordingly, wos2Δ featured significantly reduced virulence within impacting fungal burden in a murine model of cryptococcosis. Our study highlights a vulnerable point in the fungal chaperone network that offers a therapeutic opportunity to interfere with both fungal virulence and fitness.IMPORTANCEThe global impact of fungal pathogens, both emerging and emerged, is undeniable, and the alarming increase in antifungal resistance rates hampers our ability to protect the global population from deadly infections. For cryptococcal infections, a limited arsenal of antifungals and increasing rates of resistance demand alternative therapeutic strategies, including an anti-virulence approach, which disarms the pathogen of critical virulence factors, empowering the host to remove the pathogens and clear the infection. To this end, we apply state-of-the-art mass spectrometry-based proteomics to evaluate the impact of a recently defined novel co-chaperone, Wos2, toward cryptococcal virulence using in vitro and in vivo models of infection. We explore global proteome and secretome remodeling driven by the protein and uncover the novel role in modulating the fungal oxidative stress response. Complementation of proteome findings with in vitro infectivity assays demonstrated the protective role of Wos2 within the macrophage phagosome, influencing fungal replication and survival. These results underscore differential cryptococcal survivability and weakened patterns of dissemination in the absence of wos2. Overall, our study establishes Wos2 as an important contributor to fungal pathogenesis and warrants further research into critical proteins within global stress response networks as potential druggable targets to reduce fungal virulence and clear infection.
Collapse
Affiliation(s)
- Brianna Ball
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Arjun Sukumaran
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Samanta Pladwig
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Samiha Kazi
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Norris Chan
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Effie Honeywell
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Manuela Modrakova
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | | |
Collapse
|
4
|
Sagini JPN, Ligabue-Braun R. Fungal heat shock proteins: molecular phylogenetic insights into the host takeover. THE SCIENCE OF NATURE - NATURWISSENSCHAFTEN 2024; 111:16. [PMID: 38483597 DOI: 10.1007/s00114-024-01903-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/26/2024] [Accepted: 02/29/2024] [Indexed: 03/19/2024]
Abstract
Heat shock proteins are constitutively expressed chaperones induced by cellular stress, such as changes in temperature, pH, and osmolarity. These proteins, present in all organisms, are highly conserved and are recruited for the assembly of protein complexes, transport, and compartmentalization of molecules. In fungi, these proteins are related to their adaptation to the environment, their evolutionary success in acquiring new hosts, and regulation of virulence and resistance factors. These characteristics are interesting for assessment of the host adaptability and ecological transitions, given the emergence of infections by these microorganisms. Based on phylogenetic inferences, we compared the sequences of HSP9, HSP12, HSP30, HSP40, HSP70, HSP90, and HSP110 to elucidate the evolutionary relationships of different fungal organisms to suggest evolutionary patterns employing the maximum likelihood method. By the different reconstructions, our inference supports the hypothesis that these classes of proteins are associated with pathogenic gains against endothermic hosts, as well as adaptations for phytopathogenic fungi.
Collapse
Affiliation(s)
- João Pedro Nunes Sagini
- Graduate Program in Biological Sciences (PPGBio), Federal University of Health Sciences of Porto Alegre (UFCSPA), Sarmento Leite, 245, Porto Alegre, 90050-170, Brazil.
| | - Rodrigo Ligabue-Braun
- Graduate Program in Biological Sciences (PPGBio), Federal University of Health Sciences of Porto Alegre (UFCSPA), Sarmento Leite, 245, Porto Alegre, 90050-170, Brazil
- Department of Pharmacosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Sarmento Leite, 245, Porto Alegre, 90050-170, Brazil
| |
Collapse
|
5
|
Neves-da-Rocha J, Santos-Saboya MJ, Lopes MER, Rossi A, Martinez-Rossi NM. Insights and Perspectives on the Role of Proteostasis and Heat Shock Proteins in Fungal Infections. Microorganisms 2023; 11:1878. [PMID: 37630438 PMCID: PMC10456932 DOI: 10.3390/microorganisms11081878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/30/2023] [Accepted: 07/06/2023] [Indexed: 08/27/2023] Open
Abstract
Fungi are a diverse group of eukaryotic organisms that infect humans, animals, and plants. To successfully colonize their hosts, pathogenic fungi must continuously adapt to the host's unique environment, e.g., changes in temperature, pH, and nutrient availability. Appropriate protein folding, assembly, and degradation are essential for maintaining cellular homeostasis and survival under stressful conditions. Therefore, the regulation of proteostasis is crucial for fungal pathogenesis. The heat shock response (HSR) is one of the most important cellular mechanisms for maintaining proteostasis. It is activated by various stresses and regulates the activity of heat shock proteins (HSPs). As molecular chaperones, HSPs participate in the proteostatic network to control cellular protein levels by affecting their conformation, location, and degradation. In recent years, a growing body of evidence has highlighted the crucial yet understudied role of stress response circuits in fungal infections. This review explores the role of protein homeostasis and HSPs in fungal pathogenicity, including their contributions to virulence and host-pathogen interactions, as well as the concerted effects between HSPs and the main proteostasis circuits in the cell. Furthermore, we discuss perspectives in the field and the potential for targeting the components of these circuits to develop novel antifungal therapies.
Collapse
Affiliation(s)
- João Neves-da-Rocha
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil; (M.J.S.-S.); (M.E.R.L.); (A.R.)
| | | | | | | | - Nilce M. Martinez-Rossi
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil; (M.J.S.-S.); (M.E.R.L.); (A.R.)
| |
Collapse
|
6
|
Lee Y, Robbins N, Cowen LE. Molecular mechanisms governing antifungal drug resistance. NPJ ANTIMICROBIALS AND RESISTANCE 2023; 1:5. [PMID: 38686214 PMCID: PMC11057204 DOI: 10.1038/s44259-023-00007-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/17/2023] [Indexed: 05/02/2024]
Abstract
Fungal pathogens are a severe public health problem. The leading causative agents of systemic fungal infections include species from the Candida, Cryptococcus, and Aspergillus genera. As opportunistic pathogens, these fungi are generally harmless in healthy hosts; however, they can cause significant morbidity and mortality in immunocompromised patients. Despite the profound impact of pathogenic fungi on global human health, the current antifungal armamentarium is limited to only three major classes of drugs, all of which face complications, including host toxicity, unfavourable pharmacokinetics, or limited spectrum of activity. Further exacerbating this issue is the growing prevalence of antifungal-resistant infections and the emergence of multidrug-resistant pathogens. In this review, we discuss the diverse strategies employed by leading fungal pathogens to evolve antifungal resistance, including drug target alterations, enhanced drug efflux, and induction of cellular stress response pathways. Such mechanisms of resistance occur through diverse genetic alterations, including point mutations, aneuploidy formation, and epigenetic changes given the significant plasticity observed in many fungal genomes. Additionally, we highlight recent literature surrounding the mechanisms governing resistance in emerging multidrug-resistant pathogens including Candida auris and Candida glabrata. Advancing our knowledge of the molecular mechanisms by which fungi adapt to the challenge of antifungal exposure is imperative for designing therapeutic strategies to tackle the emerging threat of antifungal resistance.
Collapse
Affiliation(s)
- Yunjin Lee
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1 Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1 Canada
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1 Canada
| |
Collapse
|
7
|
de Castro Teixeira AP, Fernandes Queiroga Moraes G, de Oliveira RJ, Silva Santos C, Alves Caiana RR, Rufino de Freitas JC, Vasconcelos U, de Oliveira Pereira F, Oliveira Lima I. Antifungal Activity, Antibiofilm and Association Studies with O-Alkylamidoximes against Cryptococcus spp. Chem Biodivers 2023; 20:e202200539. [PMID: 36730650 DOI: 10.1002/cbdv.202200539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 12/23/2022] [Accepted: 02/01/2023] [Indexed: 02/04/2023]
Abstract
This is the first study that describes the antifungal and anti-biofilm potential of O-alkylamidoximes against strains of Cryptococcus neoformans and Cryptococcus gattii. In vitro tests have shown that O-alkylamidoximes are capable of inhibiting fungal growth and biofilm formation of the C. neoformans and C. gattii strains, suggesting, from molecular docking, the potential for interaction with the Hsp90. The associations between O-alkylamidoximes and amphotericin B were beneficial. Therefore, O-alkylamidoximes can be a useful alternative to contribute to the limited arsenal of drugs, since they showed a powerful action against the primary agents of Cryptococcosis.
Collapse
Affiliation(s)
- Anna Paula de Castro Teixeira
- Postgraduate Program in Natural Sciences and Biotechnology, Education and Health Center, Federal University of Campina Grande, Cuité, Brazil
| | | | | | - Cosme Silva Santos
- Postgraduate Program in Chemistry, Federal Rural University of Pernambuco, Recife, Brazil
| | - Rodrigo Ribeiro Alves Caiana
- Postgraduate Program in Natural Sciences and Biotechnology, Education and Health Center, Federal University of Campina Grande, Cuité, Brazil
| | - Juliano Carlo Rufino de Freitas
- Postgraduate Program in Natural Sciences and Biotechnology, Education and Health Center, Federal University of Campina Grande, Cuité, Brazil
- Postgraduate Program in Chemistry, Federal Rural University of Pernambuco, Recife, Brazil
| | - Ulrich Vasconcelos
- Laboratory of Animal Microbiology, Biotechnology Center, Federal University of Paraíba, João Pessoa, Brazil
| | | | - Igara Oliveira Lima
- Postgraduate Program in Natural Sciences and Biotechnology, Education and Health Center, Federal University of Campina Grande, Cuité, Brazil
| |
Collapse
|
8
|
Deng H, Song J, Huang Y, Yang C, Zang X, Zhou Y, Li H, Dai B, Xue X. Combating increased antifungal drug resistance in Cryptococcus, what should we do in the future? Acta Biochim Biophys Sin (Shanghai) 2023; 55:540-547. [PMID: 36815374 PMCID: PMC10195138 DOI: 10.3724/abbs.2023011] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/08/2022] [Indexed: 02/05/2023] Open
Abstract
Few therapeutic drugs and increased drug resistance have aggravated the current treatment difficulties of Cryptococcus in recent years. To better understand the antifungal drug resistance mechanism and treatment strategy of cryptococcosis. In this review, by combining the fundamental features of Cryptococcus reproduction leading to changes in its genome, we review recent research into the mechanism of four current anti-cryptococcal agents, coupled with new therapeutic strategies and the application of advanced technologies WGS and CRISPR-Cas9 in this field, hoping to provide a broad idea for the future clinical therapy of cryptococcosis.
Collapse
Affiliation(s)
- Hengyu Deng
- Affiliated Hospital of Weifang Medical UniversitySchool of Clinical MedicineWeifang Medical UniversityWeifang261053China
| | - Jialin Song
- Affiliated Hospital of Weifang Medical UniversitySchool of Clinical MedicineWeifang Medical UniversityWeifang261053China
| | - Yemei Huang
- of Respiratory and Critical CareBeijing Shijitan HospitalCapital Medical University; Peking University Ninth School of Clinical MedicineBeijing100089China
| | - Chen Yang
- Department of Laboratory Medicinethe First Medical CentreChinese PLA General HospitalBeijing100853China
| | - Xuelei Zang
- of Respiratory and Critical CareBeijing Shijitan HospitalCapital Medical University; Peking University Ninth School of Clinical MedicineBeijing100089China
| | - Yangyu Zhou
- of Respiratory and Critical CareBeijing Shijitan HospitalCapital Medical University; Peking University Ninth School of Clinical MedicineBeijing100089China
| | - Hongli Li
- Affiliated Hospital of Weifang Medical UniversitySchool of Clinical MedicineWeifang Medical UniversityWeifang261053China
| | - Bin Dai
- of Respiratory and Critical CareBeijing Shijitan HospitalCapital Medical University; Peking University Ninth School of Clinical MedicineBeijing100089China
| | - Xinying Xue
- Affiliated Hospital of Weifang Medical UniversitySchool of Clinical MedicineWeifang Medical UniversityWeifang261053China
- of Respiratory and Critical CareBeijing Shijitan HospitalCapital Medical University; Peking University Ninth School of Clinical MedicineBeijing100089China
| |
Collapse
|
9
|
Regulation of Hsp80 involved in the acquisition of induced thermotolerance, and NCA-2 involved in calcium stress tolerance by the calcineurin-CRZ-1 signaling pathway in Neurospora crassa. Mycol Prog 2022. [DOI: 10.1007/s11557-022-01833-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
10
|
Robbins N, Cowen LE. Antifungal discovery. Curr Opin Microbiol 2022; 69:102198. [PMID: 36037637 PMCID: PMC10726697 DOI: 10.1016/j.mib.2022.102198] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 11/30/2022]
Abstract
Fungi have a profound impact on human health, leading to billions of infections and millions of deaths worldwide each year. Exacerbating the public health burden is the continued emergence of drug-resistant fungal pathogens coupled with a dearth of treatment options to combat serious infections. Despite this health threat, scientific advances in chemistry, genetics, and biochemistry methodologies have enabled novel antifungal compounds to be discovered. Here, we describe current approaches for the discovery and characterization of novel antifungals, focusing on the identification of novel chemical matter and elucidation of the cellular target of bioactive compounds, followed by a review of the most promising emerging therapies in the antifungal-development pipeline.
Collapse
Affiliation(s)
- Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
11
|
Ngan NTT, Flower B, Day JN. Treatment of Cryptococcal Meningitis: How Have We Got Here and Where are We Going? Drugs 2022; 82:1237-1249. [PMID: 36112342 PMCID: PMC9483520 DOI: 10.1007/s40265-022-01757-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2022] [Indexed: 11/26/2022]
Abstract
Cryptococcal meningitis is a devastating brain infection cause by encapsulated yeasts of the Cryptococcus genus. Exposure, through inhalation, is likely universal by adulthood, but symptomatic infection only occurs in a minority, in most cases, months or years after exposure. Disease has been described in almost all tissues, but it is the organism’s tropism for the central nervous system that results in the most devastating illness. While invasive disease can occur in the immunocompetent, the greatest burden by far is in immunocompromised individuals, particularly people living with human immunodeficiency virus (HIV), organ transplant recipients and those on glucocorticoid therapy or other immunosuppressive drugs. Clinical presentation is variable, but diagnosis is usually straightforward, with cerebrospinal fluid microscopy, culture, and antigen testing proving significantly more sensitive than diagnostic tests for other brain infections. Although disease incidence has reduced since the advent of effective HIV therapy, mortality when disease occurs remains extremely high, and has changed little in recent decades. This Therapy in Practice review is an update of a talk first given by JND at the European Congress on Clinical Microbiology and Infectious Diseases in 2019 in the Netherlands. The review contextualizes the most recently published World Health Organization (WHO) guidelines for the treatment of HIV-associated cryptococcal meningitis in terms of the data from large, randomized, controlled trials published between 1997 and 2022. We discuss the rationale for induction and maintenance therapy and the efficacy and undesirable effects of the current therapeutic armamentarium of amphotericin, flucytosine and fluconazole. We address recent research into repurposed drugs such as sertraline and tamoxifen, and potential future treatment options, including the novel antifungals fosmanogepix, efungumab and oteseconazole, and non-pharmaceutical solutions such as neurapheresis cerebrospinal fluid filtration.
Collapse
Affiliation(s)
- Nguyen Thi Thuy Ngan
- Department of Tropical Medicine, Cho Ray Hospital, Ho Chi Minh City, Vietnam
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Barnaby Flower
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Jeremy N Day
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam.
- Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
12
|
Moghimi S, Shafiei M, Foroumadi A. Drug design strategies for the treatment azole-resistant candidiasis. Expert Opin Drug Discov 2022; 17:879-895. [PMID: 35793245 DOI: 10.1080/17460441.2022.2098949] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Despite the availability of novel antifungals and therapeutic strategies, the rate of global mortality linked to invasive fungal diseases from fungal infection remains high. Candida albicans account for the most invasive mycosis produced by yeast. Thus, the current arsenal of medicinal chemists is focused on finding new effective agents with lower toxicity and broad-spectrum activity. In this review article, recent efforts to find effective agents against azole-resistant candidiasis, a common fungal infection, are covered. AREAS COVERED Herein, the authors outlined all azole-based compounds, dual target, and new scaffolds (non-azole-based compounds) which were effective against azole-resistant candidiasis. In addition, the mechanism of action and SAR studies were also discussed, if the data were available. EXPERT OPINION The current status of fungal infections and the drawbacks of existing drugs have encouraged scientists to find novel scaffolds based on different methods like virtual screening and fragment-based drug discovery. Machine learning and in-silico methods have found their role in this field and experts are hopeful to find novel scaffolds/compounds by using these methods.
Collapse
Affiliation(s)
- Setareh Moghimi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Shafiei
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Alireza Foroumadi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Souza JAM, Gurgel ILDS, Malacco NLSDO, Martins FRB, Queiroz-Junior CM, Teixeira MM, Soriani FM. Pre-Exposure With Extracellular Vesicles From Aspergillus fumigatus Attenuates Inflammatory Response and Enhances Fungal Clearance in a Murine Model Pulmonary Aspergillosis. Front Cell Infect Microbiol 2022; 12:898619. [PMID: 35719346 PMCID: PMC9198263 DOI: 10.3389/fcimb.2022.898619] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/27/2022] [Indexed: 11/23/2022] Open
Abstract
Aspergillus fumigatus is a ubiquitous and saprophytic filamentous fungus and the main etiologic agent of aspergillosis. Infections caused by A. fumigatus culminate in a strong inflammatory response that can evolve into respiratory failure and may be lethal in immunocompromised individuals. In the last decades, it has been demonstrated that extracellular vesicles (EVs) elicit a notable biological response in immune cells. EVs carry a variety of biomolecules, therefore are considered potential antigen delivery vehicles. The role of EVs as a strategy for modulating an effective response against infections caused by A. fumigatus remains unexplored. Here we investigate the use of EVs derived from A. fumigatus as an immunization tool to induce a more robust immune response to A. fumigatus pulmonary infection. In order to investigate that, male C57BL/6 mice were immunized with two doses of EVs and infected with A. fumigatus. Pre-exposure of mice to EVs was able to induce the production of specific IgG serum for fungal antigens. Besides that, the immunization with EVs reduced the neutrophilic infiltrate into the alveoli, as well as the extravasation of total proteins and the production of proinflammatory mediators IL-1β, IL-6, and CXCL-1. In addition, immunization prevented extensive lung tissue damage and also improved phagocytosis and fungus clearance. Noteworthy, immunization with EVs, associated with subclinical doses of Amphotericin B (AmB) treatment, rescued 50% of mice infected with A. fumigatus from lethal fungal pneumonia. Therefore, the present study shows a new role for A. fumigatus EVs as host inflammatory response modulators, suggesting their use as immunizing agents.
Collapse
Affiliation(s)
- Jéssica Amanda Marques Souza
- Centro de Pesquisa e Desenvolvimento de Fármacos, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- *Correspondence: Frederico Marianetti Soriani, ; Jéssica Amanda Marques Souza,
| | - Isabella Luísa da Silva Gurgel
- Centro de Pesquisa e Desenvolvimento de Fármacos, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Nathália Luísa Sousa de Oliveira Malacco
- Centro de Pesquisa e Desenvolvimento de Fármacos, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- The Lopes Lab, Institute of Parasitology, McGill University, Montreal, QC, Canada
| | - Flávia Rayssa Braga Martins
- Centro de Pesquisa e Desenvolvimento de Fármacos, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Celso Martins Queiroz-Junior
- Centro de Pesquisa e Desenvolvimento de Fármacos, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Martins Teixeira
- Centro de Pesquisa e Desenvolvimento de Fármacos, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Frederico Marianetti Soriani
- Centro de Pesquisa e Desenvolvimento de Fármacos, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- *Correspondence: Frederico Marianetti Soriani, ; Jéssica Amanda Marques Souza,
| |
Collapse
|
14
|
Heat shock proteins and the calcineurin-crz1 signaling regulate stress responses in fungi. Arch Microbiol 2022; 204:240. [PMID: 35377020 DOI: 10.1007/s00203-022-02833-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 12/26/2022]
Abstract
The heat shock proteins (Hsps) act as a molecular chaperone to stabilize client proteins involved in various cell functions in fungi. Hsps are classified into different families such as HSP90, HSP70, HSP60, HSP40, and small HSPs (sHsps). Hsp90, a well-studied member of the Hsp family proteins, plays a role in growth, cell survival, and pathogenicity in fungi. Hsp70 and sHsps are involved in the development, tolerance to stress conditions, and drug resistance in fungi. Hsp60 is a mitochondrial chaperone, and Hsp40 regulates fungal ATPase machinery. In this review, we describe the cell functions, regulation, and the molecular link of the Hsps with the calcineurin-crz1 calcium signaling pathway for their role in cell survival, growth, virulence, and drug resistance in fungi and related organisms.
Collapse
|
15
|
Gene, virulence and related regulatory mechanisms in Cryptococcus gattii. Acta Biochim Biophys Sin (Shanghai) 2022; 54:593-603. [PMID: 35593469 PMCID: PMC9828318 DOI: 10.3724/abbs.2022029] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Cryptococcus gattii is a kind of basidiomycetous yeast, which grows in human and animal hosts. C. gattii has four distinct genomes, VGI/AFLP4, VGII/AFLP6, VGIII/AFLP5, and VGIV/AFLP7. The virulence of C. gattii is closely associated with genotype and related stress-signaling pathways, but the pathogenic mechanism of C. gattii has not been fully identified. With the development of genomics and transcriptomics, the relationship among genes, regulatory mechanisms, virulence, and treatment is gradually being recognized. In this review, to better understand how C. gattii causes disease and to characterize hypervirulent C. gattii strains, we summarize the current understanding of C. gattii genotypes, phenotypes, virulence, and the regulatory mechanisms.
Collapse
|
16
|
Fu C, Beattie SR, Jezewski AJ, Robbins N, Whitesell L, Krysan DJ, Cowen LE. Genetic analysis of Hsp90 function in Cryptococcus neoformans highlights key roles in stress tolerance and virulence. Genetics 2022; 220:iyab164. [PMID: 34849848 PMCID: PMC8733452 DOI: 10.1093/genetics/iyab164] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/27/2021] [Indexed: 11/13/2022] Open
Abstract
The opportunistic human fungal pathogen Cryptococcus neoformans has tremendous impact on global health, causing 181,000 deaths annually. Current treatment options are limited, and the frequent development of drug resistance exacerbates the challenge of managing invasive cryptococcal infections. In diverse fungal pathogens, the essential molecular chaperone Hsp90 governs fungal survival, drug resistance, and virulence. Therefore, targeting this chaperone has emerged as a promising approach to combat fungal infections. However, the role of Hsp90 in supporting C. neoformans pathogenesis remains largely elusive due to a lack of genetic characterization. To help dissect the functions of Hsp90 in C. neoformans, we generated a conditional expression strain in which HSP90 is under control of the copper-repressible promoter CTR4-2. Addition of copper to culture medium depleted Hsp90 transcript and protein levels in this strain, resulting in compromised fungal growth at host temperature; increased sensitivity to stressors, including the azole class of antifungals; altered C. neoformans morphology; and impaired melanin production. Finally, leveraging the fact that copper concentrations vary widely in different mouse tissues, we demonstrated attenuated virulence for the CTR4-2p-HSP90 mutant specifically in an inhalation model of Cryptococcus infection. During invasion and establishment of infection in this mouse model, the pathogen is exposed to the relatively high copper concentrations found in the lung as compared to blood. Overall, this work generates a tractable genetic system to study the role of Hsp90 in supporting the pathogenicity of C. neoformans and provides proof-of-principle that targeting Hsp90 holds great promise as a strategy to control cryptococcal infection.
Collapse
Affiliation(s)
- Ci Fu
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Sarah R Beattie
- Departments of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Andrew J Jezewski
- Departments of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Luke Whitesell
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Damian J Krysan
- Departments of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Microbiology/Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1M1, Canada
| |
Collapse
|
17
|
Crunden JL, Diezmann S. Hsp90 interaction networks in fungi-tools and techniques. FEMS Yeast Res 2021; 21:6413543. [PMID: 34718512 PMCID: PMC8599792 DOI: 10.1093/femsyr/foab054] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 10/26/2021] [Indexed: 01/01/2023] Open
Abstract
Heat-shock protein 90 (Hsp90) is a central regulator of cellular proteostasis. It stabilizes numerous proteins that are involved in fundamental processes of life, including cell growth, cell-cycle progression and the environmental response. In addition to stabilizing proteins, Hsp90 governs gene expression and controls the release of cryptic genetic variation. Given its central role in evolution and development, it is important to identify proteins and genes that interact with Hsp90. This requires sophisticated genetic and biochemical tools, including extensive mutant collections, suitable epitope tags, proteomics approaches and Hsp90-specific pharmacological inhibitors for chemogenomic screens. These usually only exist in model organisms, such as the yeast Saccharomyces cerevisiae. Yet, the importance of other fungal species, such as Candida albicans and Cryptococcus neoformans, as serious human pathogens accelerated the development of genetic tools to study their virulence and stress response pathways. These tools can also be exploited to map Hsp90 interaction networks. Here, we review tools and techniques for Hsp90 network mapping available in different fungi and provide a summary of existing mapping efforts. Mapping Hsp90 networks in fungal species spanning >500 million years of evolution provides a unique vantage point, allowing tracking of the evolutionary history of eukaryotic Hsp90 networks.
Collapse
Affiliation(s)
- Julia L Crunden
- School of Cellular and Molecular Medicine, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - Stephanie Diezmann
- School of Cellular and Molecular Medicine, University of Bristol, University Walk, Bristol BS8 1TD, UK
| |
Collapse
|
18
|
Horianopoulos LC, Lee CWJ, Hu G, Caza M, Kronstad JW. Dnj1 Promotes Virulence in Cryptococcus neoformans by Maintaining Robust Endoplasmic Reticulum Homeostasis Under Temperature Stress. Front Microbiol 2021; 12:727039. [PMID: 34566931 PMCID: PMC8461255 DOI: 10.3389/fmicb.2021.727039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/16/2021] [Indexed: 11/13/2022] Open
Abstract
The capacity of opportunistic fungal pathogens such as Cryptococcus neoformans to cause disease is dependent on their ability to overcome an onslaught of stresses including elevated temperature under mammalian host conditions. Protein chaperones and co-chaperones play key roles in thermotolerance. In this study, we characterized the role of the endoplasmic reticulum (ER) J-domain containing co-chaperone, Dnj1, in the virulence of C. neoformans. A strain expressing a Dnj1-GFP fusion protein was used to confirm localization to the ER, and a dnj1∆ deletion mutant was shown to be hypersensitive to the ER stress caused by tunicamycin (TM) or 4μ8C. Dnj1 and another ER chaperone, calnexin were found to coordinately maintain ER homeostasis and contribute to maintenance of cell wall architecture. Dnj1 also contributed to thermotolerance and increased in abundance at elevated temperatures representative of febrile patients (e.g., 39°C) thus highlighting its role as a temperature-responsive J domain protein. The elaboration of virulence factors such as the polysaccharide capsule and extracellular urease activity were also markedly impaired in the dnj1∆ mutant when induced at human body temperature (i.e., 37°C). These virulence factors are immunomodulatory and, indeed, infection with the dnj1∆ mutant revealed impaired induction of the cytokines IL-6, IL-10, and MCP-1 in the lungs of mice compared to infection with wild type or complemented strains. The dnj1∆ mutant also had attenuated virulence in an intranasal murine model of cryptococcosis. Altogether, our data indicate that Dnj1 is crucial for survival and virulence factor production at elevated temperatures. The characterization of this co-chaperone also highlights the importance of maintaining homeostasis in the ER for the pathogenesis of C. neoformans.
Collapse
Affiliation(s)
| | - Christopher W J Lee
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Guanggan Hu
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Mélissa Caza
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - James W Kronstad
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
19
|
Alaalm L, Crunden JL, Butcher M, Obst U, Whealy R, Williamson CE, O'Brien HE, Schaffitzel C, Ramage G, Spencer J, Diezmann S. Identification and Phenotypic Characterization of Hsp90 Phosphorylation Sites That Modulate Virulence Traits in the Major Human Fungal Pathogen Candida albicans. Front Cell Infect Microbiol 2021; 11:637836. [PMID: 34513723 PMCID: PMC8431828 DOI: 10.3389/fcimb.2021.637836] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 05/24/2021] [Indexed: 01/13/2023] Open
Abstract
The highly conserved, ubiquitous molecular chaperone Hsp90 is a key regulator of cellular proteostasis and environmental stress responses. In human pathogenic fungi, which kill more than 1.6 million patients each year worldwide, Hsp90 governs cellular morphogenesis, drug resistance, and virulence. Yet, our understanding of the regulatory mechanisms governing fungal Hsp90 function remains sparse. Post-translational modifications are powerful components of nature’s toolbox to regulate protein abundance and function. Phosphorylation in particular is critical in many cellular signaling pathways and errant phosphorylation can have dire consequences for the cell. In the case of Hsp90, phosphorylation affects its stability and governs its interactions with co-chaperones and clients. Thereby modulating the cell’s ability to cope with environmental stress. Candida albicans, one of the leading human fungal pathogens, causes ~750,000 life-threatening invasive infections worldwide with unacceptably high mortality rates. Yet, it remains unknown if and how Hsp90 phosphorylation affects C. albicans virulence traits. Here, we show that phosphorylation of Hsp90 is critical for expression of virulence traits. We combined proteomics, molecular evolution analyses and structural modeling with molecular biology to characterize the role of Hsp90 phosphorylation in this non-model pathogen. We demonstrated that phosphorylation negatively affects key virulence traits, such as the thermal stress response, morphogenesis, and drug susceptibility. Our results provide the first record of a specific Hsp90 phosphorylation site acting as modulator of fungal virulence. Post-translational modifications of Hsp90 could prove valuable in future exploitations as antifungal drug targets.
Collapse
Affiliation(s)
- Leenah Alaalm
- Department of Biology & Biochemistry, University of Bath, Bath, United Kingdom
| | - Julia L Crunden
- Department of Biology & Biochemistry, University of Bath, Bath, United Kingdom.,School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Mark Butcher
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, United Kingdom
| | - Ulrike Obst
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Ryann Whealy
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | | | - Heath E O'Brien
- MRC Centre for Neuropsychiatric Genetics & Genomics, Division of Psychological Medicine & Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | | | - Gordon Ramage
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, United Kingdom
| | - James Spencer
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Stephanie Diezmann
- Department of Biology & Biochemistry, University of Bath, Bath, United Kingdom.,School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
20
|
Brilhante RSN, Gotay WJP, Pereira VS, de Oliveira JS, Pereira-Neto WA, Castelo-Branco DDSCM, Cordeiro RDA, Sidrim JJC, Rocha MFG. Antifungal activity of promethazine and chlorpromazine against planktonic cells and biofilms of Cryptococcus neoformans/Cryptococcus gattii complex species. Med Mycol 2021; 58:906-912. [PMID: 32016364 DOI: 10.1093/mmy/myz140] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 12/22/2019] [Accepted: 01/27/2020] [Indexed: 12/14/2022] Open
Abstract
Cryptococcus neoformans/Cryptococcus gattii are fungal pathogens that affect the central nervous system, mainly in immunocompromised individuals. Due to the limited pharmacological arsenal available for the treatment of cryptococcosis associated with cases of antifungal resistance of Cryptococcus spp. reported in some studies, the search for new compounds with antifungal potential becomes relevant. Thus, the objective of this study was to evaluate the inhibitory effect of phenothiazines (promethazine and chlorpromazine) on C. neoformans/C. gattii planktonic cells and biofilms. In vitro planktonic susceptibility testing was performed using the broth microdilution assay. The effect of phenothiazines was evaluated against biofilm formation and mature Cryptococcus biofilms. Biofilm morphology and ultrastructure were also evaluated by scanning electron microscopy. Promethazine and chlorpromazine showed antifungal activity against planktonic cells, with minimum inhibitory concentrations of 8-32 μg/ml and 4-16 μg/ml, respectively. As for biofilm formation, phenothiazines reduced biomass by 60% and metabolic activity by 90% at 64 μg/ml; while in mature biofilms, reductions of 85% and 90% in biomass and metabolic activity, respectively, were observed at 1024 μg/ml. Promethazine and chlorpromazine were also able to disrupt and fragment biofilms. In conclusion, promethazine and chlorpromazine have antifungal activity against planktonic cells and biofilms of Cryptococcus spp. These data show the potential of promethazine and chlorpromazine as antibiofilm drugs.
Collapse
Affiliation(s)
- Raimunda Sâmia Nogueira Brilhante
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Wilker Jose Perez Gotay
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Vandbergue Santos Pereira
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Jonathas Sales de Oliveira
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Waldemiro Aquino Pereira-Neto
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Débora de Souza Collares Maia Castelo-Branco
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Rossana de Aguiar Cordeiro
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - José Júlio Costa Sidrim
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Marcos Fábio Gadelha Rocha
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil.,Postgraduate Program in Veterinary Sciences, College of Veterinary, State University of Ceará. Av. Dr. Silas Munguba, 1700, Campus do Itaperi, CEP: 60714-903, Fortaleza, Ceará, Brazil
| |
Collapse
|
21
|
Lee Y, Puumala E, Robbins N, Cowen LE. Antifungal Drug Resistance: Molecular Mechanisms in Candida albicans and Beyond. Chem Rev 2021; 121:3390-3411. [PMID: 32441527 PMCID: PMC8519031 DOI: 10.1021/acs.chemrev.0c00199] [Citation(s) in RCA: 433] [Impact Index Per Article: 108.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Fungal infections are a major contributor to infectious disease-related deaths across the globe. Candida species are among the most common causes of invasive mycotic disease, with Candida albicans reigning as the leading cause of invasive candidiasis. Given that fungi are eukaryotes like their human host, the number of unique molecular targets that can be exploited for antifungal development remains limited. Currently, there are only three major classes of drugs approved for the treatment of invasive mycoses, and the efficacy of these agents is compromised by the development of drug resistance in pathogen populations. Notably, the emergence of additional drug-resistant species, such as Candida auris and Candida glabrata, further threatens the limited armamentarium of antifungals available to treat these serious infections. Here, we describe our current arsenal of antifungals and elaborate on the resistance mechanisms Candida species possess that render them recalcitrant to therapeutic intervention. Finally, we highlight some of the most promising therapeutic strategies that may help combat antifungal resistance, including combination therapy, targeting fungal-virulence traits, and modulating host immunity. Overall, a thorough understanding of the mechanistic principles governing antifungal drug resistance is fundamental for the development of novel therapeutics to combat current and emerging fungal threats.
Collapse
Affiliation(s)
- Yunjin Lee
- Department of Molecular Genetics, University of Toronto, 661 University Avenue, Toronto, Ontario M5G 1M1, Canada
| | - Emily Puumala
- Department of Molecular Genetics, University of Toronto, 661 University Avenue, Toronto, Ontario M5G 1M1, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, 661 University Avenue, Toronto, Ontario M5G 1M1, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, 661 University Avenue, Toronto, Ontario M5G 1M1, Canada
| |
Collapse
|
22
|
Horianopoulos LC, Kronstad JW. Chaperone Networks in Fungal Pathogens of Humans. J Fungi (Basel) 2021; 7:209. [PMID: 33809191 PMCID: PMC7998936 DOI: 10.3390/jof7030209] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/05/2021] [Accepted: 03/10/2021] [Indexed: 12/14/2022] Open
Abstract
The heat shock proteins (HSPs) function as chaperones to facilitate proper folding and modification of proteins and are of particular importance when organisms are subjected to unfavourable conditions. The human fungal pathogens are subjected to such conditions within the context of infection as they are exposed to human body temperature as well as the host immune response. Herein, the roles of the major classes of HSPs are briefly reviewed and their known contributions in human fungal pathogens are described with a focus on Candida albicans, Cryptococcus neoformans, and Aspergillus fumigatus. The Hsp90s and Hsp70s in human fungal pathogens broadly contribute to thermotolerance, morphological changes required for virulence, and tolerance to antifungal drugs. There are also examples of J domain co-chaperones and small HSPs influencing the elaboration of virulence factors in human fungal pathogens. However, there are diverse members in these groups of chaperones and there is still much to be uncovered about their contributions to pathogenesis. These HSPs do not act in isolation, but rather they form a network with one another. Interactions between chaperones define their specific roles and enhance their protein folding capabilities. Recent efforts to characterize these HSP networks in human fungal pathogens have revealed that there are unique interactions relevant to these pathogens, particularly under stress conditions. The chaperone networks in the fungal pathogens are also emerging as key coordinators of pathogenesis and antifungal drug tolerance, suggesting that their disruption is a promising strategy for the development of antifungal therapy.
Collapse
Affiliation(s)
| | - James W. Kronstad
- Michael Smith Laboratories, Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada;
| |
Collapse
|
23
|
Exploring Small Heat Shock Proteins (sHSPs) for Targeting Drug Resistance in Candida albicans and other Pathogenic Fungi. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2021. [DOI: 10.22207/jpam.15.1.42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Fungal infections have predominantly increased worldwide that leads to morbidity and mortality in severe cases. Invasive candidiasis and other pathogenic fungal infections are a major problem in immunocompromised individuals and post-operative patients. Increasing resistance to existing antifungal drugs calls for the identification of novel antifungal drug targets for chemotherapeutic interventions. This demand for identification and characterization of novel drug targets leads to the development of effective antifungal therapy against drug resistant fungi. Heat shock proteins (HSPs) are important for various biological processes like protein folding, posttranslational modifications, transcription, translation, and protein aggregation. HSPs are involved in maintaining homeostasis of the cell. A subgroup of HSPs is small heat shock proteins (sHSPs), which functions as cellular chaperones. They are having a significant role in the many cellular functions like development, cytoskeletal organization, apoptosis, membrane lipid polymorphism, differentiation, autophagy, in infection recognition and are major players in various stresses like osmotic stress, pH stress, etc. Studies have shown that fungal cells express increased levels of sHSPs upon antifungal drug induced stress responses. Here we review the important role of small heat shock proteins (sHSPs) in fungal diseases and their potential as antifungal targets.
Collapse
|
24
|
Marcyk PT, LeBlanc EV, Kuntz DA, Xue A, Ortiz F, Trilles R, Bengtson S, Kenney TM, Huang DS, Robbins N, Williams NS, Krysan DJ, Privé GG, Whitesell L, Cowen LE, Brown LE. Fungal-Selective Resorcylate Aminopyrazole Hsp90 Inhibitors: Optimization of Whole-Cell Anticryptococcal Activity and Insights into the Structural Origins of Cryptococcal Selectivity. J Med Chem 2021; 64:1139-1169. [PMID: 33444025 PMCID: PMC8493596 DOI: 10.1021/acs.jmedchem.0c01777] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The essential eukaryotic chaperone Hsp90 regulates the form and function of diverse client proteins, many of which govern thermotolerance, virulence, and drug resistance in fungal species. However, use of Hsp90 inhibitors as antifungal therapeutics has been precluded by human host toxicities and suppression of immune responses. We recently described resorcylate aminopyrazoles (RAPs) as the first class of Hsp90 inhibitors capable of discriminating between fungal (Cryptococcus neoformans, Candida albicans) and human isoforms of Hsp90 in biochemical assays. Here, we report an iterative structure-property optimization toward RAPs capable of inhibiting C. neoformans growth in culture. In addition, we report the first X-ray crystal structures of C. neoformans Hsp90 nucleotide binding domain (NBD), as the apoprotein and in complexes with the non-species-selective Hsp90 inhibitor NVP-AUY922 and three RAPs revealing unique ligand-induced conformational rearrangements, which reaffirm the hypothesis that intrinsic differences in protein flexibility can confer selective inhibition of fungal versus human Hsp90 isoforms.
Collapse
Affiliation(s)
- Paul T. Marcyk
- Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, Boston, Massachusetts 02215, United States
| | - Emmanuelle V. LeBlanc
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Douglas A. Kuntz
- Princess Margaret Cancer Centre, Toronto, Ontario, M5G 1L7, Canada
| | - Alice Xue
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Francisco Ortiz
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas 75390-9038, United States
| | - Richard Trilles
- Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, Boston, Massachusetts 02215, United States
| | - Stephen Bengtson
- Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, Boston, Massachusetts 02215, United States
| | - Tristan M.G. Kenney
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, M5G 1L7, Canada
| | - David S. Huang
- Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, Boston, Massachusetts 02215, United States
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Noelle S. Williams
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas 75390-9038, United States
| | - Damian J. Krysan
- Departments of Pediatrics and Microbiology/Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, 52242, United States
| | - Gilbert G. Privé
- Princess Margaret Cancer Centre, Toronto, Ontario, M5G 1L7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, M5G 1L7, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Luke Whitesell
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Lauren E. Brown
- Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, Boston, Massachusetts 02215, United States
| |
Collapse
|
25
|
Treatment strategies for cryptococcal infection: challenges, advances and future outlook. Nat Rev Microbiol 2021; 19:454-466. [PMID: 33558691 PMCID: PMC7868659 DOI: 10.1038/s41579-021-00511-0] [Citation(s) in RCA: 185] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2021] [Indexed: 01/31/2023]
Abstract
Cryptococcus spp., in particular Cryptococcus neoformans and Cryptococcus gattii, have an enormous impact on human health worldwide. The global burden of cryptococcal meningitis is almost a quarter of a million cases and 181,000 deaths annually, with mortality rates of 100% if infections remain untreated. Despite these alarming statistics, treatment options for cryptococcosis remain limited, with only three major classes of drugs approved for clinical use. Exacerbating the public health burden is the fact that the only new class of antifungal drugs developed in decades, the echinocandins, displays negligible antifungal activity against Cryptococcus spp., and the efficacy of the remaining therapeutics is hampered by host toxicity and pathogen resistance. Here, we describe the current arsenal of antifungal agents and the treatment strategies employed to manage cryptococcal disease. We further elaborate on the recent advances in our understanding of the intrinsic and adaptive resistance mechanisms that are utilized by Cryptococcus spp. to evade therapeutic treatments. Finally, we review potential therapeutic strategies, including combination therapy, the targeting of virulence traits, impairing stress response pathways and modulating host immunity, to effectively treat infections caused by Cryptococcus spp. Overall, understanding of the mechanisms that regulate anti-cryptococcal drug resistance, coupled with advances in genomics technologies and high-throughput screening methodologies, will catalyse innovation and accelerate antifungal drug discovery.
Collapse
|
26
|
Carolus H, Pierson S, Lagrou K, Van Dijck P. Amphotericin B and Other Polyenes-Discovery, Clinical Use, Mode of Action and Drug Resistance. J Fungi (Basel) 2020; 6:E321. [PMID: 33261213 PMCID: PMC7724567 DOI: 10.3390/jof6040321] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/25/2020] [Accepted: 11/25/2020] [Indexed: 12/21/2022] Open
Abstract
Although polyenes were the first broad spectrum antifungal drugs on the market, after 70 years they are still the gold standard to treat a variety of fungal infections. Polyenes such as amphotericin B have a controversial image. They are the antifungal drug class with the broadest spectrum, resistance development is still relatively rare and fungicidal properties are extensive. Yet, they come with a significant host toxicity that limits their use. Relatively recently, the mode of action of polyenes has been revised, new mechanisms of drug resistance were discovered and emergent polyene resistant species such as Candida auris entered the picture. This review provides a short description of the history and clinical use of polyenes, and focusses on the ongoing debate concerning their mode of action, the diversity of resistance mechanisms discovered to date and the most recent trends in polyene resistance development.
Collapse
Affiliation(s)
- Hans Carolus
- VIB-KU Leuven Center for Microbiology, 3001 Leuven, Belgium; (H.C.); (S.P.)
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, 3001 Leuven, Belgium
| | - Siebe Pierson
- VIB-KU Leuven Center for Microbiology, 3001 Leuven, Belgium; (H.C.); (S.P.)
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, 3001 Leuven, Belgium
| | - Katrien Lagrou
- Laboratory of Clinical Bacteriology and Mycology, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3001 Leuven, Belgium;
- Department of Laboratory Medicine and National Reference Center for Mycosis, UZ Leuven, 3001 Leuven, Belgium
| | - Patrick Van Dijck
- VIB-KU Leuven Center for Microbiology, 3001 Leuven, Belgium; (H.C.); (S.P.)
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, 3001 Leuven, Belgium
| |
Collapse
|
27
|
LeBlanc EV, Polvi EJ, Veri AO, Privé GG, Cowen LE. Structure-guided approaches to targeting stress responses in human fungal pathogens. J Biol Chem 2020; 295:14458-14472. [PMID: 32796038 DOI: 10.1074/jbc.rev120.013731] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 08/11/2020] [Indexed: 11/06/2022] Open
Abstract
Fungi inhabit extraordinarily diverse ecological niches, including the human body. Invasive fungal infections have a devastating impact on human health worldwide, killing ∼1.5 million individuals annually. The majority of these deaths are attributable to species of Candida, Cryptococcus, and Aspergillus Treating fungal infections is challenging, in part due to the emergence of resistance to our limited arsenal of antifungal agents, necessitating the development of novel therapeutic options. Whereas conventional antifungal strategies target proteins or cellular components essential for fungal growth, an attractive alternative strategy involves targeting proteins that regulate fungal virulence or antifungal drug resistance, such as regulators of fungal stress responses. Stress response networks enable fungi to adapt, grow, and cause disease in humans and include regulators that are highly conserved across eukaryotes as well as those that are fungal-specific. This review highlights recent developments in elucidating crystal structures of fungal stress response regulators and emphasizes how this knowledge can guide the design of fungal-selective inhibitors. We focus on the progress that has been made with highly conserved regulators, including the molecular chaperone Hsp90, the protein phosphatase calcineurin, and the small GTPase Ras1, as well as with divergent stress response regulators, including the cell wall kinase Yck2 and trehalose synthases. Exploring structures of these important fungal stress regulators will accelerate the design of selective antifungals that can be deployed to combat life-threatening fungal diseases.
Collapse
Affiliation(s)
- Emmanuelle V LeBlanc
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Elizabeth J Polvi
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Amanda O Veri
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Gilbert G Privé
- Departments of Medical Biophysics and Biochemistry, University of Toronto, Toronto, Ontario, Canada.,Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
28
|
Brilhante RSN, Silva JAT, Araújo GDS, Pereira VS, Gotay WJP, Oliveira JSD, Guedes GMDM, Pereira-Neto WA, Castelo-Branco DDSCM, Cordeiro RDA, Sidrim JJC, Rocha MFG. Darunavir inhibits Cryptococcus neoformans/ Cryptococcus gattii species complex growth and increases the susceptibility of biofilms to antifungal drugs. J Med Microbiol 2020; 69:830-837. [PMID: 32459616 DOI: 10.1099/jmm.0.001194] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Introduction. Cryptococcus species are pathogens commonly associated with cases of meningoencephalitis in individuals who are immunosuppressed due to AIDS.Aim. The aim was to evaluate the effects of the antiretroviral darunavir alone or associated with fluconazole, 5-flucytosine and amphotericin B against planktonic cells and biofilms of Cryptococcus species.Methodology. Susceptibility testing of darunavir and the common antifungals against 12 members of the Cryptococcus neoformans/Cryptococcus gattii species complex was evaluated by broth microdilution. The interaction between darunavir and antifungals against planktonic cells was tested by a checkerboard assay. The effects of darunavir against biofilm metabolic activity and biomass were evaluated by the XTT reduction assay and crystal violet staining, respectively.Results. Darunavir combined with amphotericin B showed a synergistic interaction against planktonic cells. No antagonistic interaction was observed between darunavir and the antifungals used. All Cryptococcus species strains were strong biofilm producers. Darunavir alone reduced biofilm metabolic activity and biomass when added during and after biofilm formation (P<0.05). The combination of darunavir with antifungals caused a significant reduction in biofilm metabolic activity and biomass when compared to darunavir alone (P<0.05).Conclusion. Darunavir presents antifungal activity against planktonic cells of Cryptococcus species and synergism with amphotericin B. In addition, darunavir led to reduced biofilm formation and showed activity against mature biofilms of Cryptococcus species. Activity of the antifungals against mature biofilms was enhanced in the presence of darunavir.
Collapse
Affiliation(s)
- Raimunda Sâmia Nogueira Brilhante
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - José Alexandre Telmos Silva
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Géssica Dos Santos Araújo
- Postgraduate Program in Veterinary Sciences, College of Veterinary, State University of Ceará. Av. Dr. Silas Munguba, 1700, Campus do Itaperi, CEP: 60714-903, Fortaleza, Ceará, Brazil
| | - Vandbergue Santos Pereira
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Wilker Jose Perez Gotay
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Jonathas Sales de Oliveira
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Glaucia Morgana de Melo Guedes
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Waldemiro Aquino Pereira-Neto
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Débora de Souza Collares Maia Castelo-Branco
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Rossana de Aguiar Cordeiro
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - José Júlio Costa Sidrim
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Marcos Fábio Gadelha Rocha
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| |
Collapse
|
29
|
The Novel J-Domain Protein Mrj1 Is Required for Mitochondrial Respiration and Virulence in Cryptococcus neoformans. mBio 2020; 11:mBio.01127-20. [PMID: 32518190 PMCID: PMC7373193 DOI: 10.1128/mbio.01127-20] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cryptococcus neoformans is the causative agent of cryptococcal meningitis, a disease responsible for ∼15% of all HIV-related deaths. Unfortunately, development of antifungal drugs is challenging because potential targets are conserved between humans and C. neoformans. In this context, we characterized a unique J-domain protein, Mrj1, which lacks orthologs in humans. We showed that Mrj1 was required for normal mitochondrial respiration and that mutants lacking Mrj1 were deficient in growth, capsule elaboration, and virulence. Furthermore, we were able to phenocopy the defects in growth and capsule elaboration by inhibiting respiration. This result suggests that the role of Mrj1 in mitochondrial function was responsible for the observed virulence defects and reinforces the importance of mitochondria to fungal pathogenesis. Mitochondria are difficult to target, as their function is also key to human cells; however, Mrj1 presents an opportunity to target a unique fungal protein required for mitochondrial function and virulence in C. neoformans. The opportunistic fungal pathogen Cryptococcus neoformans must adapt to the mammalian environment to establish an infection. Proteins facilitating adaptation to novel environments, such as chaperones, may be required for virulence. In this study, we identified a novel mitochondrial co-chaperone, Mrj1 (mitochondrial respiration J-domain protein 1), necessary for virulence in C. neoformans. The mrj1Δ and J-domain-inactivated mutants had general growth defects at both routine laboratory and human body temperatures and were deficient in the major virulence factor of capsule elaboration. The latter phenotype was associated with cell wall changes and increased capsular polysaccharide shedding. Accordingly, the mrj1Δ mutant was avirulent in a murine model of cryptococcosis. Mrj1 has a mitochondrial localization and co-immunoprecipitated with Qcr2, a core component of complex III of the electron transport chain. The mrj1 mutants were deficient in mitochondrial functions, including growth on alternative carbon sources, growth without iron, and mitochondrial polarization. They were also insensitive to complex III inhibitors and hypersensitive to an alternative oxidase (AOX) inhibitor, suggesting that Mrj1 functions in respiration. In support of this conclusion, mrj1 mutants also had elevated basal oxygen consumption rates which were completely abolished by the addition of the AOX inhibitor, confirming that Mrj1 is required for mitochondrial respiration through complexes III and IV. Furthermore, inhibition of complex III phenocopied the capsule and cell wall defects of the mrj1 mutants. Taken together, these results indicate that Mrj1 is required for normal mitochondrial respiration, a key aspect of adaptation to the host environment and virulence.
Collapse
|
30
|
Spadari CDC, Wirth F, Lopes LB, Ishida K. New Approaches for Cryptococcosis Treatment. Microorganisms 2020; 8:E613. [PMID: 32340403 PMCID: PMC7232457 DOI: 10.3390/microorganisms8040613] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 02/06/2023] Open
Abstract
Cryptococcosis is an important opportunistic infection and a leading cause of meningitis in patients with HIV infection. The antifungal pharmacological treatment is limited to amphotericin B, fluconazole and 5- flucytosine. In addition to the limited pharmacological options, the high toxicity, increased resistance rate and difficulty of the currently available antifungal molecules to cross the blood-brain barrier hamper the treatment. Thus, the search for new alternatives for the treatment of cryptococcal meningitis is extremely necessary. In this review, we describe the therapeutic strategies currently available, discuss new molecules with antifungal potential in different phases of clinical trials and in advanced pre-clinical phase, and examine drug nanocarriers to improve delivery to the central nervous system.
Collapse
Affiliation(s)
- Cristina de Castro Spadari
- Laboratory of Antifungal Chemotherapy, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (C.d.C.S.); (F.W.)
| | - Fernanda Wirth
- Laboratory of Antifungal Chemotherapy, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (C.d.C.S.); (F.W.)
| | - Luciana Biagini Lopes
- Laboratory of Nanomedicine and Drug Delivery Systems, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil;
| | - Kelly Ishida
- Laboratory of Antifungal Chemotherapy, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (C.d.C.S.); (F.W.)
| |
Collapse
|
31
|
Huang DS, LeBlanc EV, Shekhar-Guturja T, Robbins N, Krysan DJ, Pizarro J, Whitesell L, Cowen LE, Brown LE. Design and Synthesis of Fungal-Selective Resorcylate Aminopyrazole Hsp90 Inhibitors. J Med Chem 2020; 63:2139-2180. [PMID: 31513387 PMCID: PMC7069776 DOI: 10.1021/acs.jmedchem.9b00826] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The molecular chaperone Hsp90, essential in all eukaryotes, plays a multifaceted role in promoting survival, virulence, and drug resistance across diverse pathogenic fungal species. The chaperone is also critically important, however, to the pathogen's human host, preventing the use of known clinical Hsp90 inhibitors in antifungal applications due to concomitant host toxicity issues. With the goal of developing Hsp90 inhibitors with acceptable therapeutic indices for the treatment of invasive fungal infections, we initiated a program to design and synthesize potent inhibitors with selective activity against fungal Hsp90 isoforms over their human counterparts. Building on our previously reported derivatization of resorcylate natural products to produce fungal-selective compounds, we have developed a series of synthetic aminopyrazole-substituted resorcylate amides with broad, potent, and fungal-selective Hsp90 inhibitory activity. Herein we describe the synthesis of this series, as well as biochemical structure-activity relationships driving selectivity for the Hsp90 isoforms expressed by Cryptococcus neoformans and Candida albicans, two pathogenic fungi of major clinical importance.
Collapse
Affiliation(s)
- David S. Huang
- Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, Boston, MA, 02215, USA
| | - Emmanuelle V. LeBlanc
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Tanvi Shekhar-Guturja
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Damian J. Krysan
- Departments of Pediatrics and Microbiology/Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Juan Pizarro
- Department of Tropical Medicine, School of Public Health and Tropical Medicine and Vector-Borne Infectious Disease Research Center, Tulane University, New Orleans, LA, 70112, USA
| | - Luke Whitesell
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Lauren E. Brown
- Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, Boston, MA, 02215, USA
| |
Collapse
|
32
|
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: 16] [Impact Index Per Article: 3.2] [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
|
33
|
Oshiro KGN, Rodrigues G, Monges BED, Cardoso MH, Franco OL. Bioactive Peptides Against Fungal Biofilms. Front Microbiol 2019; 10:2169. [PMID: 31681179 PMCID: PMC6797862 DOI: 10.3389/fmicb.2019.02169] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 09/04/2019] [Indexed: 12/20/2022] Open
Abstract
Infections caused by invasive fungal biofilms have been widely associated with high morbidity and mortality rates, mainly due to the advent of antibiotic resistance. Moreover, fungal biofilms impose an additional challenge, leading to multidrug resistance. This fact, along with the contamination of medical devices and the limited number of effective antifungal agents available on the market, demonstrates the importance of finding novel drug candidates targeting pathogenic fungal cells and biofilms. In this context, an alternative strategy is the use of antifungal peptides (AFPs) against fungal biofilms. AFPs are considered a group of bioactive molecules with broad-spectrum activities and multiple mechanisms of action that have been widely used as template molecules for drug design strategies aiming at greater specificity and biological efficacy. Among the AFP classes most studied in the context of fungal biofilms, defensins, cathelicidins and histatins have been described. AFPs can also act by preventing the formation of fungal biofilms and eradicating preformed biofilms through mechanisms associated with cell wall perturbation, inhibition of planktonic fungal cells’ adhesion onto surfaces, gene regulation and generation of reactive oxygen species (ROS). Thus, considering the critical scenario imposed by fungal biofilms and associated infections and the application of AFPs as a possible treatment, this review will focus on the most effective AFPs described to date, with a core focus on antibiofilm peptides, as well as their efficacy in vivo, application on surfaces and proposed mechanisms of action.
Collapse
Affiliation(s)
- Karen G N Oshiro
- Programa de Pós-Graduação em Patologia Molecular, Faculdade de Medicina, Universidade de Brasília, Brasília, Brazil.,S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
| | - Gisele Rodrigues
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brazil
| | - Bruna Estéfani D Monges
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
| | - Marlon Henrique Cardoso
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil.,Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brazil
| | - Octávio Luiz Franco
- Programa de Pós-Graduação em Patologia Molecular, Faculdade de Medicina, Universidade de Brasília, Brasília, Brazil.,S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil.,Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brazil
| |
Collapse
|
34
|
Souza JAM, Baltazar LDM, Carregal VM, Gouveia-Eufrasio L, de Oliveira AG, Dias WG, Campos Rocha M, Rocha de Miranda K, Malavazi I, Santos DDA, Frézard FJG, de Souza DDG, Teixeira MM, Soriani FM. Characterization of Aspergillus fumigatus Extracellular Vesicles and Their Effects on Macrophages and Neutrophils Functions. Front Microbiol 2019; 10:2008. [PMID: 31551957 PMCID: PMC6738167 DOI: 10.3389/fmicb.2019.02008] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 08/16/2019] [Indexed: 12/26/2022] Open
Abstract
Extracellular vesicles (EVs) has been considered an alternative process for intercellular communication. EVs release by filamentous fungi and the role of vesicular secretion during fungus-host cells interaction remain unknown. Here, we identified the secretion of EVs from the pathogenic filamentous fungus, Aspergillus fumigatus. Analysis of the structure of EVs demonstrated that A. fumigatus produces round shaped bilayer structures ranging from 100 to 200 nm size, containing ergosterol and a myriad of proteins involved in REDOX, cell wall remodeling and metabolic functions of the fungus. We demonstrated that macrophages can phagocytose A. fumigatus EVs. Phagocytic cells, stimulated with EVs, increased fungal clearance after A. fumigatus conidia challenge. EVs were also able to induce the production of TNF-α and CCL2 by macrophages and a synergistic effect was observed in the production of these mediators when the cells were challenged with the conidia. In bone marrow-derived neutrophils (BMDN) treated with EVs, there was enhancement of the production of TNF-α and IL-1β in response to conidia. Together, our results demonstrate, for the first time, that A. fumigatus produces EVs containing a diverse set of proteins involved in fungal physiology and virulence. Moreover, EVs are biologically active and stimulate production of inflammatory mediators and fungal clearance.
Collapse
Affiliation(s)
- Jéssica Amanda Marques Souza
- Centro de Pesquisa e Desenvolvimento de Fármacos, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ludmila de Matos Baltazar
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Virgínia Mendes Carregal
- Laboratório de Biofísica e Sistemas Nanoestruturados, Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ludmila Gouveia-Eufrasio
- Laboratório de Micologia, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - André Gustavo de Oliveira
- Lab Circuitos Fisiológicos, Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Wendell Girard Dias
- Plataforma de Microscopia Eletrônica Rudolf Barth, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Marina Campos Rocha
- Centro de Ciências Biológicas e da Saúde, Departamento de Genética e Evolução, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Kildare Rocha de Miranda
- Laboratório de Ultraestrutura Celular Hertha Meyer, Programa de Biologia Celular e Parasitologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Iran Malavazi
- Centro de Ciências Biológicas e da Saúde, Departamento de Genética e Evolução, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Daniel de Assis Santos
- Laboratório de Micologia, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Frédéric Jean Georges Frézard
- Laboratório de Biofísica e Sistemas Nanoestruturados, Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Daniele da Glória de Souza
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Martins Teixeira
- Centro de Pesquisa e Desenvolvimento de Fármacos, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Frederico Marianetti Soriani
- Centro de Pesquisa e Desenvolvimento de Fármacos, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
35
|
Jia C, Zhang J, Zhuge Y, Xu K, Liu J, Wang J, Li L, Chu M. Synergistic effects of geldanamycin with fluconazole are associated with reactive oxygen species in Candida tropicalis resistant to azoles and amphotericin B. Free Radic Res 2019; 53:618-628. [PMID: 31185751 DOI: 10.1080/10715762.2019.1610563] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Chang Jia
- Pediatric Research Institute, the Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, PR China
| | - Jian Zhang
- Children’s Heart Center, Institute of Cardiovascular Development and Translational Medicine, the Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, PR China
| | - Yingzhi Zhuge
- Children’s Heart Center, Institute of Cardiovascular Development and Translational Medicine, the Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, PR China
| | - Ke Xu
- Institute of Life Sciences, Wenzhou University, Wenzhou, PR China
| | - Jiahui Liu
- Department of Clinical Laboratory, the Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, PR China
| | - Jinle Wang
- Department of Clinical Laboratory, the Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, PR China
| | - Lei Li
- School of Medicine, Wenzhou Key Laboratory of Cardiovascular Development and Translational Medicine, the Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, PR China
| | - Maoping Chu
- Pediatric Research Institute, the Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, PR China
- Children’s Heart Center, Institute of Cardiovascular Development and Translational Medicine, the Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, PR China
| |
Collapse
|
36
|
Monk BC, Sagatova AA, Hosseini P, Ruma YN, Wilson RK, Keniya MV. Fungal Lanosterol 14α-demethylase: A target for next-generation antifungal design. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2019; 1868:140206. [PMID: 30851431 DOI: 10.1016/j.bbapap.2019.02.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 02/15/2019] [Accepted: 02/21/2019] [Indexed: 12/19/2022]
Abstract
The cytochrome P450 enzyme lanosterol 14α-demethylase (LDM) is the target of the azole antifungals used widely in medicine and agriculture as prophylaxis or treatments of infections or diseases caused by fungal pathogens. These drugs and agrochemicals contain an imidazole, triazole or tetrazole substituent, with one of the nitrogens in the azole ring coordinating as the sixth axial ligand to the LDM heme iron. Structural studies show that this membrane bound enzyme contains a relatively rigid ligand binding pocket comprised of a deeply buried heme-containing active site together with a substrate entry channel and putative product exit channel that reach to the membrane. Within the ligand binding pocket the azole antifungals have additional affinity determining interactions with hydrophobic side-chains, the polypeptide backbone and via water-mediated hydrogen bond networks. This review will describe the tools that can be used to identify and characterise the next generation of antifungals targeting LDM, with the goal of obtaining highly potent broad-spectrum fungicides that will be able to avoid target and drug efflux mediated antifungal resistance.
Collapse
Affiliation(s)
- Brian C Monk
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, PO Box 56, Dunedin 9054, New Zealand.
| | - Alia A Sagatova
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Parham Hosseini
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Yasmeen N Ruma
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Rajni K Wilson
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Mikhail V Keniya
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| |
Collapse
|
37
|
Rocha MC, Santos CA, Malavazi I. The Regulatory Function of the Molecular Chaperone Hsp90 in the Cell Wall Integrity of Pathogenic Fungi. CURR PROTEOMICS 2018. [DOI: 10.2174/1570164615666180820155807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Different signaling cascades including the Cell Wall Integrity (CWI), the High Osmolarity Glycerol (HOG) and the Ca2+/calcineurin pathways control the cell wall biosynthesis and remodeling in fungi. Pathogenic fungi, such as Aspergillus fumigatus and Candida albicans, greatly rely on these signaling circuits to cope with different sources of stress, including the cell wall stress evoked by antifungal drugs and the host’s response during infection. Hsp90 has been proposed as an important regulatory protein and an attractive target for antifungal therapy since it stabilizes major effector proteins that act in the CWI, HOG and Ca2+/calcineurin pathways. Data from the human pathogen C. albicans have provided solid evidence that loss-of-function of Hsp90 impairs the evolution of resistance to azoles and echinocandin drugs. In A. fumigatus, Hsp90 is also required for cell wall integrity maintenance, reinforcing a coordinated function of the CWI pathway and this essential molecular chaperone. In this review, we focus on the current information about how Hsp90 impacts the aforementioned signaling pathways and consequently the homeostasis and maintenance of the cell wall, highlighting this cellular event as a key mechanism underlying antifungal therapy based on Hsp90 inhibition.
Collapse
Affiliation(s)
- Marina Campos Rocha
- Departmento de Genetica e Evolucao, Centro de Ciencias Biologicas e da Saude, Universidade Federal de Sao Carlos, Sao Carlos, Brazil
| | - Camilla Alves Santos
- Departmento de Genetica e Evolucao, Centro de Ciencias Biologicas e da Saude, Universidade Federal de Sao Carlos, Sao Carlos, Brazil
| | - Iran Malavazi
- Departmento de Genetica e Evolucao, Centro de Ciencias Biologicas e da Saude, Universidade Federal de Sao Carlos, Sao Carlos, Brazil
| |
Collapse
|
38
|
Van Dijck P, Sjollema J, Cammue BPA, Lagrou K, Berman J, d’Enfert C, Andes DR, Arendrup MC, Brakhage AA, Calderone R, Cantón E, Coenye T, Cos P, Cowen LE, Edgerton M, Espinel-Ingroff A, Filler SG, Ghannoum M, Gow NA, Haas H, Jabra-Rizk MA, Johnson EM, Lockhart SR, Lopez-Ribot JL, Maertens J, Munro CA, Nett JE, Nobile CJ, Pfaller MA, Ramage G, Sanglard D, Sanguinetti M, Spriet I, Verweij PE, Warris A, Wauters J, Yeaman MR, Zaat SA, Thevissen K. Methodologies for in vitro and in vivo evaluation of efficacy of antifungal and antibiofilm agents and surface coatings against fungal biofilms. MICROBIAL CELL (GRAZ, AUSTRIA) 2018; 5:300-326. [PMID: 29992128 PMCID: PMC6035839 DOI: 10.15698/mic2018.07.638] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 05/24/2018] [Indexed: 12/13/2022]
Abstract
Unlike superficial fungal infections of the skin and nails, which are the most common fungal diseases in humans, invasive fungal infections carry high morbidity and mortality, particularly those associated with biofilm formation on indwelling medical devices. Therapeutic management of these complex diseases is often complicated by the rise in resistance to the commonly used antifungal agents. Therefore, the availability of accurate susceptibility testing methods for determining antifungal resistance, as well as discovery of novel antifungal and antibiofilm agents, are key priorities in medical mycology research. To direct advancements in this field, here we present an overview of the methods currently available for determining (i) the susceptibility or resistance of fungal isolates or biofilms to antifungal or antibiofilm compounds and compound combinations; (ii) the in vivo efficacy of antifungal and antibiofilm compounds and compound combinations; and (iii) the in vitro and in vivo performance of anti-infective coatings and materials to prevent fungal biofilm-based infections.
Collapse
Affiliation(s)
- Patrick Van Dijck
- VIB-KU Leuven Center for Microbiology, Leuven, Belgium
- KU Leuven Laboratory of Molecular Cell Biology, Leuven, Belgium
| | - Jelmer Sjollema
- University of Groningen, University Medical Center Groningen, Department of BioMedical Engineering, Groningen, The Netherlands
| | - Bruno P. A. Cammue
- Centre for Microbial and Plant Genetics, KU Leuven, Leuven, Belgium
- Department of Plant Systems Biology, VIB, Ghent, Belgium
| | - Katrien Lagrou
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
- Clinical Department of Laboratory Medicine and National Reference Center for Mycosis, UZ Leuven, Belgium
| | - Judith Berman
- School of Molecular Cell Biology and Biotechnology, Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Israel
| | - Christophe d’Enfert
- Institut Pasteur, INRA, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - David R. Andes
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Maiken C. Arendrup
- Unit of Mycology, Statens Serum Institut, Copenhagen, Denmark
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Axel A. Brakhage
- Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute (HKI), Dept. Microbiology and Molecular Biology, Friedrich Schiller University Jena, Institute of Microbiology, Jena, Germany
| | - Richard Calderone
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington DC, USA
| | - Emilia Cantón
- Severe Infection Research Group: Medical Research Institute La Fe (IISLaFe), Valencia, Spain
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
- ESCMID Study Group for Biofilms, Switzerland
| | - Paul Cos
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Belgium
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Mira Edgerton
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY USA
| | | | - Scott G. Filler
- Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Mahmoud Ghannoum
- Center for Medical Mycology, Department of Dermatology, University Hospitals Cleveland Medical Center and Case Western Re-serve University, Cleveland, OH, USA
| | - Neil A.R. Gow
- MRC Centre for Medical Mycology, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Hubertus Haas
- Biocenter - Division of Molecular Biology, Medical University Innsbruck, Innsbruck, Austria
| | - Mary Ann Jabra-Rizk
- Department of Oncology and Diagnostic Sciences, School of Dentistry; Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, USA
| | - Elizabeth M. Johnson
- National Infection Service, Public Health England, Mycology Reference Laboratory, Bristol, UK
| | | | | | - Johan Maertens
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium and Clinical Department of Haematology, UZ Leuven, Leuven, Belgium
| | - Carol A. Munro
- MRC Centre for Medical Mycology, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Jeniel E. Nett
- University of Wisconsin-Madison, Departments of Medicine and Medical Microbiology & Immunology, Madison, WI, USA
| | - Clarissa J. Nobile
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, USA
| | - Michael A. Pfaller
- Departments of Pathology and Epidemiology, University of Iowa, Iowa, USA
- JMI Laboratories, North Liberty, Iowa, USA
| | - Gordon Ramage
- ESCMID Study Group for Biofilms, Switzerland
- College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Dominique Sanglard
- Institute of Microbiology, University of Lausanne and University Hospital, CH-1011 Lausanne
| | - Maurizio Sanguinetti
- Institute of Microbiology, Università Cattolica del Sacro Cuore, IRCCS-Fondazione Policlinico "Agostino Gemelli", Rome, Italy
| | - Isabel Spriet
- Pharmacy Dpt, University Hospitals Leuven and Clinical Pharmacology and Pharmacotherapy, Dpt. of Pharmaceutical and Pharma-cological Sciences, KU Leuven, Belgium
| | - Paul E. Verweij
- Center of Expertise in Mycology Radboudumc/CWZ, Radboud University Medical Center, Nijmegen, the Netherlands (omit "Nijmegen" in Radboud University Medical Center)
| | - Adilia Warris
- MRC Centre for Medical Mycology, Aberdeen Fungal Group, University of Aberdeen, Foresterhill, Aberdeen, UK
| | - Joost Wauters
- KU Leuven-University of Leuven, University Hospitals Leuven, Department of General Internal Medicine, Herestraat 49, B-3000 Leuven, Belgium
| | - Michael R. Yeaman
- Geffen School of Medicine at the University of California, Los Angeles, Divisions of Molecular Medicine & Infectious Diseases, Har-bor-UCLA Medical Center, LABioMed at Harbor-UCLA Medical Center
| | - Sebastian A.J. Zaat
- Department of Medical Microbiology, Amsterdam Infection and Immunity Institute, Academic Medical Center, University of Am-sterdam, Netherlands
| | - Karin Thevissen
- Centre for Microbial and Plant Genetics, KU Leuven, Leuven, Belgium
| |
Collapse
|
39
|
Aguiar Cordeiro RD, de Jesus Evangelista AJ, Serpa R, Colares de Andrade AR, Leite Mendes PB, Silva Franco JD, de Oliveira JS, de Alencar LP, Sales JA, Carneiro Câmara LM, Souza Collares Maia Castelo-Branco DD, Nogueira Brilhante RS, Costa Sidrim JJ, Gadelha Rocha MF. β-lactam antibiotics & vancomycin increase the growth & virulence of Candida spp. Future Microbiol 2018; 13:869-875. [PMID: 29882422 DOI: 10.2217/fmb-2018-0019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To investigate the direct effect of antibiotics on growth and virulence of the major Candida species associated with invasive infections. MATERIALS & METHODS Cefepime, imipenem, meropenem, amoxicillin and vancomycin were tested at twofold the peak plasma concentration (2× PP) and the peak plasma concentration (PP). The effects of antibiotics on Candida albicans, Candida parapsilosis, Candida krusei and Candida tropicalis were investigated by colony counting, flow cytometry, proteolytic activity and virulence in Caenorhabditis elegans. RESULTS Antibiotics increase growth and proteolytic activity of Candida spp; In addition, amoxicillin potentiates virulence of C. krusei and C. tropicalis against Caenorhabditis elegans. CONCLUSION These results suggest that antimicrobial therapy may have a direct effect on the pathophysiology of invasive fungal infections in patients at risk.
Collapse
Affiliation(s)
- Rossana de Aguiar Cordeiro
- Department of Pathology & Legal Medicine, Medical Mycology Specialized Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Antonio José de Jesus Evangelista
- Department of Pathology & Legal Medicine, Medical Mycology Specialized Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Rosana Serpa
- Departament of of Plant Protection, Federal University of Pelotas, Rio Grande do Sul, Brazil
| | - Ana Raquel Colares de Andrade
- Department of Pathology & Legal Medicine, Medical Mycology Specialized Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Patrícia Bruna Leite Mendes
- Department of Pathology & Legal Medicine, Medical Mycology Specialized Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Jônatas da Silva Franco
- Department of Pathology & Legal Medicine, Medical Mycology Specialized Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Jonathas Sales de Oliveira
- Department of Pathology & Legal Medicine, Medical Mycology Specialized Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Lucas Pereira de Alencar
- Department of Pathology & Legal Medicine, Medical Mycology Specialized Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Jamille Alencar Sales
- Department of Pathology & Legal Medicine, Medical Mycology Specialized Center, Federal University of Ceará, Fortaleza, Ceará, Brazil.,Department of Veterinary Medicine, Post Graduate Program in Veterinary Sciences, College of Veterinary Medicine, State University of Ceará, Fortaleza, Ceará, Brazil
| | - Lília Maria Carneiro Câmara
- Department of Pathology & Legal Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | | | - Raimunda Sâmia Nogueira Brilhante
- Department of Pathology & Legal Medicine, Medical Mycology Specialized Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - José Júlio Costa Sidrim
- Department of Pathology & Legal Medicine, Medical Mycology Specialized Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Marcos Fábio Gadelha Rocha
- Department of Pathology & Legal Medicine, Medical Mycology Specialized Center, Federal University of Ceará, Fortaleza, Ceará, Brazil.,Department of Veterinary Medicine, Post Graduate Program in Veterinary Sciences, College of Veterinary Medicine, State University of Ceará, Fortaleza, Ceará, Brazil
| |
Collapse
|
40
|
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
|
41
|
Xu L, Li Y, Biggins JB, Bowman BR, Verdine GL, Gloer JB, Alspaugh JA, Bills GF. Identification of cyclosporin C from Amphichorda felina using a Cryptococcus neoformans differential temperature sensitivity assay. Appl Microbiol Biotechnol 2018; 102:2337-2350. [PMID: 29396588 DOI: 10.1007/s00253-018-8792-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/02/2018] [Accepted: 01/16/2018] [Indexed: 12/25/2022]
Abstract
We used a temperature differential assay with the opportunistic fungal pathogen Cryptococcus neoformans as a simple screening platform to detect small molecules with antifungal activity in natural product extracts. By screening of a collection extracts from two different strains of the coprophilous fungus, Amphichorda felina, we detected strong, temperature-dependent antifungal activity using a two-plate agar zone of inhibition assay at 25 and 37 °C. Bioassay-guided fractionation of the crude extract followed by liquid chromatography-mass spectrometry (LC-MS) and nuclear magnetic resonance spectroscopy (NMR) identified cyclosporin C (CsC) as the main component of the crude extract responsible for growth inhibition of C. neoformans at 37 °C. The presence of CsC was confirmed by comparison with a commercial standard. We sequenced the genome of A. felina to identify and annotate the CsC biosynthetic gene cluster. The only previously characterized gene cluster for the biosynthesis of similar compounds is that of the related immunosuppressant drug cyclosporine A (CsA). The CsA and CsC gene clusters share a high degree of synteny and sequence similarity. Amino acid changes in the adenylation domain of the CsC nonribosomal peptide synthase's sixth module may be responsible for the substitution of L-threonine compared to L-α-aminobutyric acid in the CsA peptide core. This screening strategy promises to yield additional antifungal natural products with a focused spectrum of antimicrobial activity.
Collapse
Affiliation(s)
- Lijian Xu
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, 1881 East Road, 3SCR6.4676, Houston, TX, 77054, USA
- College of Agricultural Resources and Environment, Heilongjiang University, Harbin, 150080, China
| | - Yan Li
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, 1881 East Road, 3SCR6.4676, Houston, TX, 77054, USA
- Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - John B Biggins
- LifeMine Therapeutics, 430 E. 29th Street, Suite 830, New York, NY, 10016, USA
| | - Brian R Bowman
- LifeMine Therapeutics, 430 E. 29th Street, Suite 830, New York, NY, 10016, USA
| | - Gregory L Verdine
- LifeMine Therapeutics, 430 E. 29th Street, Suite 830, New York, NY, 10016, USA
| | - James B Gloer
- Department of Chemistry, University of Iowa, Iowa City, IA, 52242, USA
| | - J Andrew Alspaugh
- Departments of Biochemistry and Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Gerald F Bills
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, 1881 East Road, 3SCR6.4676, Houston, TX, 77054, USA.
| |
Collapse
|
42
|
Gong Y, Li T, Yu C, Sun S. Candida albicans Heat Shock Proteins and Hsps-Associated Signaling Pathways as Potential Antifungal Targets. Front Cell Infect Microbiol 2017; 7:520. [PMID: 29312897 PMCID: PMC5742142 DOI: 10.3389/fcimb.2017.00520] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 12/07/2017] [Indexed: 11/28/2022] Open
Abstract
In recent decades, the incidence of invasive fungal infections has increased notably. Candida albicans (C. albicans), a common opportunistic fungal pathogen that dwells on human mucosal surfaces, can cause fungal infections, especially in immunocompromised and high-risk surgical patients. In addition, the wide use of antifungal agents has likely contributed to resistance of C. albicans to traditional antifungal drugs, increasing the difficulty of treatment. Thus, it is urgent to identify novel antifungal drugs to cope with C. albicans infections. Heat shock proteins (Hsps) exist in most organisms and are expressed in response to thermal stress. In C. albicans, Hsps control basic physiological activities or virulence via interaction with a variety of diverse regulators of cellular signaling pathways. Moreover, it has been demonstrated that Hsps confer drug resistance to C. albicans. Many studies have shown that disrupting the normal functions of C. albicans Hsps inhibits fungal growth or reverses the tolerance of C. albicans to traditional antifungal drugs. Here, we review known functions of the diverse Hsp family, Hsp-associated intracellular signaling pathways and potential antifungal targets based on these pathways in C. albicans. We hope this review will aid in revealing potential new roles of C. albicans Hsps in addition to canonical heat stress adaptions and provide more insight into identifying potential novel antifungal targets.
Collapse
Affiliation(s)
- Ying Gong
- School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Tao Li
- Intensive Care Unit, Qianfoshan Hospital Affiliated to Shandong University, Jinan, China
| | - Cuixiang Yu
- Respiration Medicine, Qianfoshan Hospital Affiliated to Shandong University, Jinan, China
| | - Shujuan Sun
- Department of Pharmacy, Qianfoshan Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
43
|
Heat shock protein 90 localizes to the surface and augments virulence factors of Cryptococcus neoformans. PLoS Negl Trop Dis 2017; 11:e0005836. [PMID: 28783748 PMCID: PMC5559104 DOI: 10.1371/journal.pntd.0005836] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 08/16/2017] [Accepted: 07/26/2017] [Indexed: 12/15/2022] Open
Abstract
Background Thermotolerance is an essential attribute for pathogenesis of Cryptococcus as exemplified by the fact that only two species in the genus, which can grow at 37°C, are human pathogens. Species which have other virulence factors including capsule formation and melanisation, but lack the ability to propagate at 37°C are not pathogenic. In another related fungal pathogen, Candida albicans, heat shock protein 90 has been implicated to be a central player in commanding pathogenicity by governing yeast to hyphal transition and drug resistance. Exploring Hsp90 biology in Cryptococcus in context of thermotolerance may thus highlight important regulatory principles of virulence and open new therapeutic avenues. Methodology/Principal findings Hsp90 is involved in regulating thermotolerance in Cryptococcus as indicated by growth hypersensitivity at 37°C upon mild compromise of Hsp90 function relative to 25°C. Biochemical studies revealed a more potent inhibition of ATPase activity by pharmacological inhibitor 17-AAG at 37°C as compared to 25°C. Catalytic efficiency of the protein at 37°C was found to be 6.39×10−5μM-1. Furthermore, indirect immunofluorescence analysis using a specific antibody revealed cell surface localization of Hsp90 via ER Golgi classical secretory pathway. Hsp90 was found to be induced under capsule inducing conditions and Hsp90 inhibition led to decrease in capsular volume. Finally compromising Hsp90 function improved anidulafungin tolerance in Cryptococcus. Conclusions/Significance Our findings highlight that Hsp90 regulates pathogenicity of the fungus by myriad ways. Firstly, it is involved in mediating thermotolerance which implies targeting Hsp90 can abrogate thermotolerance and hence growth of the fungus. Secondly, this study provides the first report of biochemical properties of Hsp90 of a pathogenic fungus. Finally, since Hsp90 is localised at the cell wall, targeting cell surface Hsp90 can represent a novel strategy to combat this lethal infection. Thermotolerance is a pre-requisite for microbes to propagate successfully as human pathogens. In this study, we have investigated the role of Heat shock protein 90 in the pathogenesis and thermotolerance of C. neoformans, an environmental fungus that causes meningoencephalitis in humans. We show that thermotolerance of Cryptococcus critically depends on Hsp90 function as modest inhibition of Hsp90 function, robustly compromised growth of the fungus at 37°C with little effect at 25°C. This observation correlated with the fact that pharmacological inhibitor, 17-AAG also showed a more potent inhibition of ATPase activity of the protein at 37°C as indicated by a lower IC50 as compared to 25°C. Indirect immunofluorescence analysis using an antibody specific to CnHsp90 revealed cell surface localization of Hsp90. BFA sensitivity of such surface localization indicated involvement of ER-Golgi classical secretory pathway for this localization. Furthermore, inhibition of Hsp90 function not only abrogated the natural resistance of C. neoformans to cell wall targeting inhibitors echinocandins but also led to decrease in capsular assembly which is one of the classical virulence determinants of the pathogen. In all, this study provides the first detailed biochemical as well as functional insights into the role of Hsp90 in governing thermotolerance and augmenting virulence factors in C. neoformans.
Collapse
|
44
|
Resistance to antifungal therapies. Essays Biochem 2017; 61:157-166. [DOI: 10.1042/ebc20160067] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/02/2017] [Accepted: 01/05/2017] [Indexed: 11/17/2022]
Abstract
The evolution of antifungal resistance among fungal pathogens has rendered the limited arsenal of antifungal drugs futile. Considering the recent rise in the number of nosocomial fungal infections in immunocompromised patients, the emerging clinical multidrug resistance (MDR) has become a matter of grave concern for medical professionals. Despite advances in therapeutic interventions, it has not yet been possible to devise convincing strategies to combat antifungal resistance. Comprehensive understanding of the molecular mechanisms of antifungal resistance is essential for identification of novel targets that do not promote or delay emergence of drug resistance. The present study discusses features and limitations of the currently available antifungals, mechanisms of antifungal resistance and highlights the emerging therapeutic strategies that could be deployed to combat MDR.
Collapse
|
45
|
Azevedo RVDM, Rizzo J, Rodrigues ML. Virulence Factors as Targets for Anticryptococcal Therapy. J Fungi (Basel) 2016; 2:jof2040029. [PMID: 29376946 PMCID: PMC5715936 DOI: 10.3390/jof2040029] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 11/18/2016] [Accepted: 11/25/2016] [Indexed: 12/24/2022] Open
Abstract
The global mortality due to cryptococcosis caused by Cryptococcus neoformans or C. gattii is unacceptably high. Currently available therapies are decades old and may be impacted by drug resistance. Therefore, the need for more effective antifungal drugs for cryptococcosis is evident. A number of Cryptococcus virulence factors have been studied in detail, providing crucial information about the fungal biology and putative molecular targets for antifungals. This review focuses on the use of well-described virulence factors of Cryptococcus as potential anticryptococcal agents.
Collapse
Affiliation(s)
- Renata V D M Azevedo
- Fundação Oswaldo Cruz-Fiocruz, Centro de Desenvolvimento Tecnológico em Saúde (CDTS), 21040-361 Rio de Janeiro, Brazil.
| | - Juliana Rizzo
- Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.
- Instituto de Bioquímica Médica (IBqM), Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.
| | - Marcio L Rodrigues
- Fundação Oswaldo Cruz-Fiocruz, Centro de Desenvolvimento Tecnológico em Saúde (CDTS), 21040-361 Rio de Janeiro, Brazil.
- Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.
| |
Collapse
|
46
|
Abstract
ABSTRACT
Invasive fungal infections are becoming an increasingly important cause of human mortality and morbidity, particularly for immunocompromised populations. The fungal pathogens
Candida albicans
,
Cryptococcus neoformans
, and
Aspergillus fumigatus
collectively contribute to over 1 million human deaths annually. Hence, the importance of safe and effective antifungal therapeutics for the practice of modern medicine has never been greater. Given that fungi are eukaryotes like their human host, the number of unique molecular targets that can be exploited for drug development remains limited. Only three classes of molecules are currently approved for the treatment of invasive mycoses. The efficacy of these agents is compromised by host toxicity, fungistatic activity, or the emergence of drug resistance in pathogen populations. Here we describe our current arsenal of antifungals and highlight current strategies that are being employed to improve the therapeutic safety and efficacy of these drugs. We discuss state-of-the-art approaches to discover novel chemical matter with antifungal activity and highlight some of the most promising new targets for antifungal drug development. We feature the benefits of combination therapy as a strategy to expand our current repertoire of antifungals and discuss the antifungal combinations that have shown the greatest potential for clinical development. Despite the paucity of new classes of antifungals that have come to market in recent years, it is clear that by leveraging innovative approaches to drug discovery and cultivating collaborations between academia and industry, there is great potential to bolster the antifungal armamentarium.
Collapse
|
47
|
Spitzer M, Robbins N, Wright GD. Combinatorial strategies for combating invasive fungal infections. Virulence 2016; 8:169-185. [PMID: 27268286 DOI: 10.1080/21505594.2016.1196300] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Invasive fungal infections are an important cause of human mortality and morbidity, particularly for immunocompromised populations. However, there remains a paucity of antifungal drug treatments available to combat these fungal pathogens. Further, antifungal compounds are plagued with problems such as host toxicity, fungistatic activity, and the emergence of drug resistance in pathogen populations. A promising therapeutic strategy to increase drug effectiveness and mitigate the emergence of drug resistance is through the use of combination drug therapy. In this review we describe the current arsenal of antifungals in medicine and elaborate on the benefits of combination therapy to expand our current antifungal drug repertoire. We examine those antifungal combinations that have shown potential against fungal pathogens and discuss strategies being employed to discover novel combination therapeutics, in particular combining antifungal agents with non-antifungal bioactive compounds. The findings summarized in this review highlight the promise of combinatorial strategies in combatting invasive mycoses.
Collapse
Affiliation(s)
- Michaela Spitzer
- a Michael G. DeGroote Institute for Infectious Disease Research and the Department of Biochemistry and Biomedical Sciences , McMaster University , Hamilton , ON , Canada
| | - Nicole Robbins
- a Michael G. DeGroote Institute for Infectious Disease Research and the Department of Biochemistry and Biomedical Sciences , McMaster University , Hamilton , ON , Canada
| | - Gerard D Wright
- a Michael G. DeGroote Institute for Infectious Disease Research and the Department of Biochemistry and Biomedical Sciences , McMaster University , Hamilton , ON , Canada
| |
Collapse
|
48
|
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
|