1
|
Yang C, Li G, Zhang Q, Bai W, Li Q, Zhang P, Zhang J. Histone deacetylase Sir2 promotes the systemic Candida albicans infection by facilitating its immune escape via remodeling the cell wall and maintaining the metabolic activity. mBio 2024; 15:e0044524. [PMID: 38682948 PMCID: PMC11237532 DOI: 10.1128/mbio.00445-24] [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: 02/14/2024] [Accepted: 03/26/2024] [Indexed: 05/01/2024] Open
Abstract
Histone deacetylation affects Candida albicans (C. albicans) pathogenicity by modulating virulence factor expression and DNA damage. The histone deacetylase Sir2 is associated with C. albicans plasticity and maintains genome stability to help C. albicans adapt to various environmental niches. However, whether Sir2-mediated chromatin modification affects C. albicans virulence is unclear. The purpose of our study was to investigate the effect of Sir2 on C. albicans pathogenicity and regulation. Here, we report that Sir2 is required for C. albicans pathogenicity, as its deletion affects the survival rate, fungal burden in different organs and the extent of tissue damage in a mouse model of disseminated candidiasis. We evaluated the impact of Sir2 on C. albicans virulence factors and revealed that the Sir2 null mutant had an impaired ability to adhere to host cells and was more easily recognized by the innate immune system. Comprehensive analysis revealed that the disruption of C. albicans adhesion was due to a decrease in cell surface hydrophobicity rather than the differential expression of adhesion genes on the cell wall. In addition, Sir2 affects the distribution and exposure of mannan and β-glucan on the cell wall, indicating that Sir2 plays a role in preventing the immune system from recognizing C. albicans. Interestingly, our results also indicated that Sir2 helps C. albicans maintain metabolic activity under hypoxic conditions, suggesting that Sir2 contributes to C. albicans colonization at hypoxic sites. In conclusion, our findings provide detailed insights into antifungal targets and a useful foundation for the development of antifungal drugs. IMPORTANCE Candida albicans (C. albicans) is the most common opportunistic fungal pathogen and can cause various superficial infections and even life-threatening systemic infections. To successfully propagate infection, this organism relies on the ability to express virulence-associated factors and escape host immunity. In this study, we demonstrated that the histone deacetylase Sir2 helps C. albicans adhere to host cells and escape host immunity by mediating cell wall remodeling; as a result, C. albicans successfully colonized and invaded the host in vivo. In addition, we found that Sir2 contributes to carbon utilization under hypoxic conditions, suggesting that Sir2 is important for C. albicans survival and the establishment of infection in hypoxic environments. In summary, we investigated the role of Sir2 in regulating C. albicans pathogenicity in detail; these findings provide a potential target for the development of antifungal drugs.
Collapse
Affiliation(s)
- Chen Yang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Pharmaceutical Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Guanglin Li
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Pharmaceutical Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qiyue Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Pharmaceutical Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Wenhui Bai
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Pharmaceutical Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qingiqng Li
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Pharmaceutical Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Peipei Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Pharmaceutical Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jiye Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Pharmaceutical Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
2
|
Drummond RA, Desai JV, Ricotta EE, Swamydas M, Deming C, Conlan S, Quinones M, Matei-Rascu V, Sherif L, Lecky D, Lee CCR, Green NM, Collins N, Zelazny AM, Prevots DR, Bending D, Withers D, Belkaid Y, Segre JA, Lionakis MS. Long-term antibiotic exposure promotes mortality after systemic fungal infection by driving lymphocyte dysfunction and systemic escape of commensal bacteria. Cell Host Microbe 2022; 30:1020-1033.e6. [PMID: 35568028 PMCID: PMC9283303 DOI: 10.1016/j.chom.2022.04.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 03/08/2022] [Accepted: 04/20/2022] [Indexed: 12/15/2022]
Abstract
Antibiotics are a modifiable iatrogenic risk factor for the most common human nosocomial fungal infection, invasive candidiasis, yet the underlying mechanisms remain elusive. We found that antibiotics enhanced the susceptibility to murine invasive candidiasis due to impaired lymphocyte-dependent IL-17A- and GM-CSF-mediated antifungal immunity within the gut. This led to non-inflammatory bacterial escape and systemic bacterial co-infection, which could be ameliorated by IL-17A or GM-CSF immunotherapy. Vancomycin alone similarly enhanced the susceptibility to invasive fungal infection and systemic bacterial co-infection. Mechanistically, vancomycin reduced the frequency of gut Th17 cells associated with impaired proliferation and RORγt expression. Vancomycin's effects on Th17 cells were indirect, manifesting only in vivo in the presence of dysbiosis. In humans, antibiotics were associated with an increased risk of invasive candidiasis and death after invasive candidiasis. Our work highlights the importance of antibiotic stewardship in protecting vulnerable patients from life-threatening infections and provides mechanistic insights into a controllable iatrogenic risk factor for invasive candidiasis.
Collapse
Affiliation(s)
- Rebecca A Drummond
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy & Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA; Institute of Immunology & Immunotherapy, Institute of Microbiology & Infection, University of Birmingham, Birmingham, B15 2TT, UK.
| | - Jigar V Desai
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy & Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Emily E Ricotta
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy & Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Muthulekha Swamydas
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy & Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Clay Deming
- Microbial Genomics Section, Translational and Functional Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Sean Conlan
- Microbial Genomics Section, Translational and Functional Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Mariam Quinones
- Bioinformatics and Computational Bioscience Branch, NIAID, NIH, Bethesda, MD 20892, USA
| | - Veronika Matei-Rascu
- Institute of Immunology & Immunotherapy, Institute of Microbiology & Infection, University of Birmingham, Birmingham, B15 2TT, UK
| | - Lozan Sherif
- Institute of Immunology & Immunotherapy, Institute of Microbiology & Infection, University of Birmingham, Birmingham, B15 2TT, UK
| | - David Lecky
- Institute of Immunology & Immunotherapy, Institute of Microbiology & Infection, University of Birmingham, Birmingham, B15 2TT, UK
| | - Chyi-Chia R Lee
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Nathaniel M Green
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy & Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Nicholas Collins
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, NIAID, NIH, Bethesda, MD 20892, USA
| | - Adrian M Zelazny
- Department of Laboratory Medicine, NIH Clinical Center, NIH, Bethesda, MD 20892, USA
| | - D Rebecca Prevots
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy & Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - David Bending
- Institute of Immunology & Immunotherapy, Institute of Microbiology & Infection, University of Birmingham, Birmingham, B15 2TT, UK
| | - David Withers
- Institute of Immunology & Immunotherapy, Institute of Microbiology & Infection, University of Birmingham, Birmingham, B15 2TT, UK
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, NIAID, NIH, Bethesda, MD 20892, USA; NIAID Microbiome Program, NIAID, NIH, Bethesda, MD 20892, USA
| | - Julia A Segre
- Microbial Genomics Section, Translational and Functional Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy & Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
3
|
Ren T, Zhu H, Tian L, Yu Q, Li M. Candida albicans infection disturbs the redox homeostasis system and induces reactive oxygen species accumulation for epithelial cell death. FEMS Yeast Res 2021; 20:5643898. [PMID: 31769804 DOI: 10.1093/femsyr/foz081] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 11/25/2019] [Indexed: 02/07/2023] Open
Abstract
Candida albicans is a common pathogenic fungus with high mortality in immunocompromised patients. However, the mechanism by which C. albicans invades host epithelial cells and causes serious tissue damage remains to be further investigated. In this study, we established the C. albicans-293T renal epithelial cell interaction model to investigate the mechanism of epithelial infection by this pathogen. It was found that C. albicans infection causes severe cell death and reactive oxygen species (ROS) accumulation in epithelial cells. Further investigations revealed that C. albicans infection might up-regulate expression of nicotinamide adenine dinucleotide phosphate (NAPDH) oxidase (NOX), inhibit the activity of the antioxidant enzymes superoxide dismutase (SOD) and catalase (CAT), and suppress the p38-Nrf2-heme oxygenase-1 (HO-1) pathway which plays an important role in the elimination of intracellular ROS. Furthermore, epithelial cell death caused by the fungal infection could be strikingly alleviated by addition of the antioxidant agent glutathione, indicating the critical role of ROS accumulation in cell death caused by the fungus. This study revealed that disturbance of the redox homeostasis system and ROS accumulation in epithelial cells is involved in cell death caused by C. albicans infection, which sheds light on the application of antioxidants in the suppression of tissue damage caused by fungal infection.
Collapse
Affiliation(s)
- Tongtong Ren
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Science, Nankai University, Tianjin 300071, P. R. China
| | - Hangqi Zhu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Science, Nankai University, Tianjin 300071, P. R. China
| | - Lei Tian
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Science, Nankai University, Tianjin 300071, P. R. China
| | - Qilin Yu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Science, Nankai University, Tianjin 300071, P. R. China
| | - Mingchun Li
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Science, Nankai University, Tianjin 300071, P. R. China
| |
Collapse
|
4
|
Teo YJ, Ng SL, Mak KW, Setiagani YA, Chen Q, Nair SK, Sheng J, Ruedl C. Renal CD169 ++ resident macrophages are crucial for protection against acute systemic candidiasis. Life Sci Alliance 2021; 4:e202000890. [PMID: 33608410 PMCID: PMC7918719 DOI: 10.26508/lsa.202000890] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 01/07/2021] [Accepted: 01/22/2021] [Indexed: 12/17/2022] Open
Abstract
Disseminated candidiasis remains as the most common hospital-acquired bloodstream fungal infection with up to 40% mortality rate despite the advancement of medical and hygienic practices. While it is well established that this infection heavily relies on the innate immune response for host survival, much less is known for the protective role elicited by the tissue-resident macrophage (TRM) subsets in the kidney, the prime organ for Candida persistence. Here, we describe a unique CD169++ TRM subset that controls Candida growth and inflammation during acute systemic candidiasis. Their absence causes severe fungal-mediated renal pathology. CD169++ TRMs, without being actively involved in direct fungal clearance, increase host resistance by promoting IFN-γ release and neutrophil ROS activity.
Collapse
Affiliation(s)
- Yi Juan Teo
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - See Liang Ng
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Keng Wai Mak
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | | | - Qi Chen
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Sajith Kumar Nair
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Jianpeng Sheng
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Christiane Ruedl
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
5
|
Edwards JE, Schwartz MM, Schmidt CS, Sobel JD, Nyirjesy P, Schodel F, Marchus E, Lizakowski M, DeMontigny EA, Hoeg J, Holmberg T, Cooke MT, Hoover K, Edwards L, Jacobs M, Sussman S, Augenbraun M, Drusano M, Yeaman MR, Ibrahim AS, Filler SG, Hennessey JP. A Fungal Immunotherapeutic Vaccine (NDV-3A) for Treatment of Recurrent Vulvovaginal Candidiasis-A Phase 2 Randomized, Double-Blind, Placebo-Controlled Trial. Clin Infect Dis 2019; 66:1928-1936. [PMID: 29697768 DOI: 10.1093/cid/ciy185] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 03/01/2018] [Indexed: 11/12/2022] Open
Abstract
Background Recurrent vulvovaginal candidiasis (RVVC) is a problematic form of mucosal Candida infection, characterized by repeated episodes per year. Candida albicans is the most common cause of RVVC. Currently, there are no immunotherapeutic treatments for RVVC. Methods This exploratory randomized, double-blind, placebo-controlled trial evaluated an immunotherapeutic vaccine (NDV-3A) containing a recombinant C. albicans adhesin/invasin protein for prevention of RVVC. Results The study in 188 women with RVVC (n = 178 evaluable) showed that 1 intramuscular dose of NDV-3A was safe and generated rapid and robust B- and T-cell immune responses. Post hoc exploratory analyses revealed a statistically significant increase in the percentage of symptom-free patients at 12 months after vaccination (42% vaccinated vs 22% placebo; P = .03) and a doubling in median time to first symptomatic episode (210 days vaccinated vs 105 days placebo) for the subset of patients aged <40 years (n = 137). The analysis of evaluable patients, which combined patients aged <40 years (77%) and ≥40 years (23%), trended toward a positive impact of NDV-3A versus placebo (P = .099). Conclusions In this unprecedented study of the effectiveness of a fungal vaccine in humans, NDV-3A administered to women with RVVC was safe and highly immunogenic and reduced the frequency of symptomatic episodes of vulvovaginal candidiasis for up to 12 months in women aged <40 years. These results support further development of NDV-3A vaccine and provide guidance for meaningful clinical endpoints for immunotherapeutic management of RVVC. Clinical Trials Registration NCT01926028.
Collapse
Affiliation(s)
- John E Edwards
- Los Angeles Biomedical Research Institute, Torrance.,David Geffen School of Medicine at University of California, Los Angeles
| | | | | | - Jack D Sobel
- School of Medicine, Wayne State University, Detroit, Michigan
| | - Paul Nyirjesy
- Drexel University College of Medicine, Philadelphia, Pennsylvania
| | | | | | | | | | - Jesse Hoeg
- NovaDigm Therapeutics, Inc, Boston, Massachusetts
| | | | | | | | - Lance Edwards
- Suffolk Obstetrics & Gynecology, Port Jefferson, New York
| | | | - Steven Sussman
- Lawrence OB-GYN Clinical Research, LLC, Lawrenceville, New Jersey
| | | | | | - Michael R Yeaman
- Los Angeles Biomedical Research Institute, Torrance.,David Geffen School of Medicine at University of California, Los Angeles
| | - Ashraf S Ibrahim
- Los Angeles Biomedical Research Institute, Torrance.,David Geffen School of Medicine at University of California, Los Angeles
| | - Scott G Filler
- Los Angeles Biomedical Research Institute, Torrance.,David Geffen School of Medicine at University of California, Los Angeles
| | | |
Collapse
|
6
|
Song Y, Li S, Zhao Y, Zhang Y, Lv Y, Jiang Y, Wang Y, Li D, Zhang H. ADH1 promotes Candida albicans pathogenicity by stimulating oxidative phosphorylation. Int J Med Microbiol 2019; 309:151330. [DOI: 10.1016/j.ijmm.2019.151330] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 06/24/2019] [Accepted: 07/09/2019] [Indexed: 01/06/2023] Open
|
7
|
Swidergall M. Candida albicans at Host Barrier Sites: Pattern Recognition Receptors and Beyond. Pathogens 2019; 8:E40. [PMID: 30934602 PMCID: PMC6471378 DOI: 10.3390/pathogens8010040] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 03/19/2019] [Accepted: 03/21/2019] [Indexed: 12/29/2022] Open
Abstract
Over the last decades, fungal infections have emerged as a growing threat to human health. Although the human body is at potential risk, various body sites host several commensal fungal species, including Candida albicans. In healthy individuals, C. albicans colonizes different mucosal surfaces without causing harm, while under diverse circumstances the fungus can proliferate and cause disease. In this context, the understanding of host⁻C. albicans interactions in health and during infection may lead to novel therapeutic approaches. Importantly, host cells express pattern recognition receptors (PRRs), which sense conserved fungal structures and orchestrate innate immune responses. Herein, important findings on the topic of the recognition of C. albicans at host barrier sites are discussed. This review briefly summarizes the importance and functions of myeloid PRRs, reviews the fungal recognition and biology of stromal cells, and highlights important C. albicans virulence attributes during site-specific proliferation and invasion.
Collapse
Affiliation(s)
- Marc Swidergall
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA 90502, USA.
- Institute for Infection and Immunity, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA.
| |
Collapse
|
8
|
Li SX, Wu HT, Liu YT, Jiang YY, Zhang YS, Liu WD, Zhu KJ, Li DM, Zhang H. The F 1F o-ATP Synthase β Subunit Is Required for Candida albicans Pathogenicity Due to Its Role in Carbon Flexibility. Front Microbiol 2018; 9:1025. [PMID: 29875745 PMCID: PMC5974098 DOI: 10.3389/fmicb.2018.01025] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 05/01/2018] [Indexed: 11/13/2022] Open
Abstract
Previous work has explored link between mitochondrial biology and fungal pathogenicity in F1Fo-ATP synthase in Candida albicans. In this work we have detailed the more specific roles of the F1Fo-ATP synthase β subunit, a key protein subunit of F1Fo-ATP synthase. The ability to assimilate alternative carbons in glucose-limited host niches is known to be a critical factor for infection caused by opportunistic pathogens including C. albicans. The function of the F1Fo-ATP synthase β subunit was characterized through the construction of an ATP2 gene null mutant (atp2Δ/Δ) and the gene-reconstituted strain (atp2Δ/ATP2) in order to understand the link between carbon metabolism and C. albicans pathogenesis. Cell growth, viability, cellular ATP content, mitochondrial membrane potential (ΔΨm), and intracellular ROS were compared between null mutant and control strain. Results showed that growth of the atp2Δ/Δ mutant in synthetic medium was slower than in complex medium. However, the synthetic medium delayed the onset of reduced cell viability and kept cellular ATP content from becoming fully depleted. Consistent with these observations, we identified transcriptional changes in metabolic response that activated other ATP-generating pathways, thereby improving cell viability during the initial phase. Unlike glucose effects, the atp2Δ/Δ mutant exhibited an immediate and sharp reduction in cell viability on non-fermentable carbon sources, consistent with an immediate depletion of cellular ATP content. Along with a reduced viability in non-fermentable carbon sources, the atp2Δ/Δ mutant displayed avirulence in a murine model of disseminated candidiasis as well as lower fungal loads in mouse organs. Regardless of the medium, however, a decrease in mitochondrial membrane potential (ΔΨm) was found in the atp2Δ/Δ mutant but ROS levels remained in the normal range. These results suggest that the F1Fo-ATP synthase β subunit is required for C. albicans pathogenicity and operates by affecting metabolic flexibility in carbon consumption.
Collapse
Affiliation(s)
- Shui-Xiu Li
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China.,Institute of Mycology, Jinan University, Guangzhou, China
| | - Hao-Tian Wu
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China.,Institute of Mycology, Jinan University, Guangzhou, China
| | - Yu-Ting Liu
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China.,Institute of Mycology, Jinan University, Guangzhou, China
| | - Yi-Ying Jiang
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China.,Institute of Mycology, Jinan University, Guangzhou, China
| | - Yi-Shan Zhang
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China.,Institute of Mycology, Jinan University, Guangzhou, China
| | - Wei-Da Liu
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences, Nanjing, China
| | - Kun-Ju Zhu
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China.,Institute of Mycology, Jinan University, Guangzhou, China
| | - Dong-Mei Li
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, DC, United States
| | - Hong Zhang
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China.,Institute of Mycology, Jinan University, Guangzhou, China
| |
Collapse
|
9
|
Organ-specific mechanisms linking innate and adaptive antifungal immunity. Semin Cell Dev Biol 2018; 89:78-90. [PMID: 29366628 DOI: 10.1016/j.semcdb.2018.01.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/09/2018] [Accepted: 01/15/2018] [Indexed: 12/24/2022]
Abstract
Fungal infections remain a significant global health problem in humans. Fungi infect millions of people worldwide and cause from acute superficial infections to life-threatening systemic disease to chronic illnesses. Trying to decipher the complex innate and adaptive immune mechanisms that protect humans from pathogenic fungi is therefore a key research goal that may lead to immune-based therapeutic strategies and improved patient outcomes. In this review, we summarize how the cells and molecules of the innate immune system activate the adaptive immune system to elicit long-term immunity to fungi. We present current knowledge and exciting new advances in the context of organ-specific immunity, outlining the tissue-specific tropisms for the major pathogenic fungi of humans, the antifungal functions of tissue-resident myeloid cells, and the adaptive immune responses required to protect specific organs from fungal challenge.
Collapse
|
10
|
Li SX, Song YJ, Zhang YS, Wu HT, Guo H, Zhu KJ, Li DM, Zhang H. Mitochondrial Complex V α Subunit Is Critical for Candida albicans Pathogenicity through Modulating Multiple Virulence Properties. Front Microbiol 2017; 8:285. [PMID: 28280492 PMCID: PMC5322696 DOI: 10.3389/fmicb.2017.00285] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Accepted: 02/13/2017] [Indexed: 11/22/2022] Open
Abstract
The α subunit (ATP1) is a vital component of mitochondrial complex V which counts for the majority of cellular ATP production in a living organism. Nevertheless, how the α subunit influences other cellular processes such as pathogenicity in Candida albicans remains poorly understood. To address this question, ATP1 mutant (atp1Δ/Δ) and the gene-reconstituted strain (atp1Δ/ATP1) have been constructed in this study and their pathogenicity-related traits are compared to those of wild type (WT). In a murine model of disseminated candidiasis, atp1Δ/Δ infected mice have a significantly higher survival rate and experience a lower fungal burden in tissues. In in vitro studies atp1Δ/Δ lose a capability to damage or destroy macrophages and endothelial cells. Furthermore, atp1Δ/Δ is not able to grow under either glucose-denial conditions or high H2O2 conditions, both of which are associated with the potency of the macrophages to kill C. albicans. Defects in filamentation and biofilm formation may impair the ability of atp1Δ/Δ to penetrate host cells and establish robust colonies in the host tissues. In concert with these pathogenic features, intracellular ATP levels of atp1Δ/Δ can drop to 1/3 of WT level. These results indicate that the α subunit of Complex V play important roles in C. albicans pathogenicity.
Collapse
Affiliation(s)
- Shui-Xiu Li
- The First Affiliated Hospital of Jinan UniversityGuangzhou, China; Institute of Mycology, Jinan UniversityGuangzhou, China
| | - Yan-Jun Song
- The First Affiliated Hospital of Jinan UniversityGuangzhou, China; Institute of Mycology, Jinan UniversityGuangzhou, China
| | - Yi-Shan Zhang
- The First Affiliated Hospital of Jinan UniversityGuangzhou, China; Institute of Mycology, Jinan UniversityGuangzhou, China
| | - Hao-Tian Wu
- The First Affiliated Hospital of Jinan UniversityGuangzhou, China; Institute of Mycology, Jinan UniversityGuangzhou, China
| | - Hui Guo
- The First Affiliated Hospital of Jinan UniversityGuangzhou, China; Institute of Mycology, Jinan UniversityGuangzhou, China
| | - Kun-Ju Zhu
- The First Affiliated Hospital of Jinan UniversityGuangzhou, China; Institute of Mycology, Jinan UniversityGuangzhou, China
| | - Dong-Mei Li
- Department of Microbiology and Immunology, Georgetown University Medical Center Washington, DC, USA
| | - Hong Zhang
- The First Affiliated Hospital of Jinan UniversityGuangzhou, China; Institute of Mycology, Jinan UniversityGuangzhou, China
| |
Collapse
|
11
|
Glass KA, Longley SJ, Bliss JM, Shaw SK. Protection of Candida parapsilosis from neutrophil killing through internalization by human endothelial cells. Virulence 2016; 6:504-14. [PMID: 26039751 DOI: 10.1080/21505594.2015.1042643] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Candida parapsilosis is a fungal pathogen that is associated with hematogenously disseminated disease in premature neonates, acutely ill or immunocompromised patients. In cell culture, C. parapsilosis cells are actively and avidly endocytosed by endothelial cells via actin polymerization mediated by N-WASP. Here we present evidence that C. parapsilosis that were internalized by endothelial cells remained alive, and avoided being acidified or otherwise damaged via the host cell. Internalized fungal cells reproduced intracellularly and eventually burst out of the host endothelial cell. When neutrophils were added to endothelium and C. parapsilosis, they patrolled the endothelial surface and efficiently killed most adherent fungal cells prior to endocytosis. But after endocytosis by endothelial cells, internalized fungal cells evaded neutrophil killing. Silencing endothelial N-WASP blocked endocytosis of C. parapsilosis and left fungal cells stranded on the cell surface, where they were susceptible to neutrophil killing. These observations suggest that for C. parapsilosis to escape from the bloodstream, fungi may adhere to and be internalized by endothelial cells before being confronted and phagocytosed by a patrolling leukocyte. Once internalized by endothelial cells, C. parapsilosis may safely replicate to cause further rounds of infection. Immunosurveillance of the intravascular lumen by leukocytes crawling on the endothelial surface and rapid killing of adherent yeast may play a major role in controlling C. parapsilosis dissemination and infected endothelial cells may be a significant reservoir for fungal persistence.
Collapse
Affiliation(s)
- Kyle A Glass
- a Department of Pediatrics; Women & Infants Hospital of Rhode Island ; Providence , RI , USA
| | | | | | | |
Collapse
|
12
|
Influence of IgG Subclass on Human Antimannan Antibody-Mediated Resistance to Hematogenously Disseminated Candidiasis in Mice. Infect Immun 2015; 84:386-94. [PMID: 26573736 DOI: 10.1128/iai.00890-15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 11/06/2015] [Indexed: 01/23/2023] Open
Abstract
Candida albicans is a yeast-like pathogen and can cause life-threatening systemic candidiasis. Its cell surface is enriched with mannan that is resistant to complement activation. Previously, we developed the recombinant human IgG1 antimannan antibody M1g1. M1g1 was found to promote complement activation and phagocytosis and protect mice from systemic candidiasis. Here, we evaluate the influence of IgG subclass on antimannan antibody-mediated protection. Three IgG subclass variants of M1g1 were constructed: M1g2, M1g3, and M1g4. The IgG subclass identity for each variant was confirmed with DNA sequence and subclass-specific antibodies. These variants contain identical M1 Fabs and exhibited similar binding affinities for C. albicans yeast and purified mannan. Yeast cells and hyphae recovered from the kidney of antibody-treated mice with systemic candidiasis showed uniform binding of each variant, indicating constitutive expression of the M1 epitope and antibody opsonization in the kidney. All variants promoted deposition of both murine and human C3 onto the yeast cell surface, with M1g4 showing delayed activation, as determined by flow cytometry and immunofluorescence microscopy. M1g4-mediated complement activation was found to be associated with its M1 Fab that activates the alternative pathway in an Fc-independent manner. Treatment with each subclass variant extended the survival of mice with systemic candidiasis (P < 0.001). However, treatment with M1g1, M1g3, or M1g4, but not with M1g2, also reduced the kidney fungal burden (P < 0.001). Thus, the role of human antimannan antibody in host resistance to systemic candidiasis is influenced by its IgG subclass.
Collapse
|
13
|
Pierce CG, Chaturvedi AK, Lazzell AL, Powell AT, Saville SP, McHardy SF, Lopez-Ribot JL. A Novel Small Molecule Inhibitor of Candida albicans Biofilm Formation, Filamentation and Virulence with Low Potential for the Development of Resistance. NPJ Biofilms Microbiomes 2015; 1. [PMID: 26691764 PMCID: PMC4681527 DOI: 10.1038/npjbiofilms.2015.12] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND/OBJECTIVES Candida albicans is the principal causative agent of candidiasis, the most common fungal infection in humans. Candidiasis represents the third-to-fourth most frequent nosocomial infection worldwide, as this normal commensal of humans causes opportunistic infections in an expanding population of immune- and medically-compromised patients. These infections are frequently associated with biofilm formation, which complicates treatment and contributes to unacceptably high mortality rates. METHODS To address the pressing need for new antifungals we have performed a high content screen of 20,000 small molecules in a chemical library (NOVACore™) to identify compounds that inhibit C. albicans biofilm formation, and conducted a series of follow-up studies to examine the in vitro and in vivo activity of the identified compounds. RESULTS The screen identified a novel series of diazaspiro-decane structural analogs which were largely represented among the bioactive compounds. Characterization of the leading compound from this series indicated that it inhibits processes associated with C. albicans virulence, most notably biofilm formation and filamentation, without having an effect on overall growth or eliciting resistance. This compound demonstrated in vivo activity in clinically-relevant murine models of both invasive and oral candidiasis and as such represents a promising lead for antifungal drug development. Furthermore, these results provide proof of concept for the implementation of anti-virulence approaches against C. albicans and other fungal infections that would be less likely to foster the emergence of resistance.
Collapse
Affiliation(s)
- Christopher G Pierce
- Department of Biology and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, United States of America
| | - Ashok K Chaturvedi
- Department of Biology and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, United States of America
| | - Anna L Lazzell
- Department of Biology and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, United States of America
| | - Alexander T Powell
- Department of Biology and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, United States of America
| | - Stephen P Saville
- Department of Biology and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, United States of America
| | - Stanton F McHardy
- Department of Chemistry and Center for Innovation in Drug Discovery, The University of Texas at San Antonio, San Antonio, TX 78249, United States of America
| | - Jose L Lopez-Ribot
- Department of Biology and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, United States of America
| |
Collapse
|
14
|
Treviño-Rangel RDJ, González GM, Martínez-Castilla AM, García-Juárez J, Robledo-Leal ER, González JG, Rosas-Taraco AG. Candida parapsilosis complex induces local inflammatory cytokines in immunocompetent mice. Med Mycol 2015; 53:612-21. [PMID: 25908650 DOI: 10.1093/mmy/myv021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 02/22/2015] [Indexed: 12/23/2022] Open
Abstract
Despite the increasing incidence of the Candida parapsilosis complex in the clinical setting and high mortality rates associated with disseminated infection, the host-fungus interactions regarding Candida parapsilosis sensu stricto and the closely related species C. orthopsilosis and C. metapsilosis remains blurred. In this study, we analyzed inflammatory cytokines levels and histopathology as well as fungal burden in spleen, kidney and lung of mice infected with six strains of the "psilosis" group with different enzymatic profiles. Strong interleukin 22 (IL-22) and tumor necrosis factor α (TNF-α) responses were observed in analyzed organs from infected mice (P < .0001) regardless of the species and enzymatic profile. TNF-α and IL-22 levels were related with spleen inflammation and fungal load. Fungal cells were detected only in spleen and kidney of infected mice, especially by day 2 post-challenge. The kidney showed glomerular retraction and partial destruction of renal tubules. Our data suggest that a strong inflammatory response, mainly of IL-22 and TNF-α, could be involved in Candida parapsilosis complex infection control.
Collapse
Affiliation(s)
| | - Gloria M González
- Departamento de Microbiología, Facultad de Medicina, Universidad Autónoma de Nuevo León. Nuevo León, Mexico
| | - Azalia M Martínez-Castilla
- Departamento de Inmunología, Facultad de Medicina, Universidad Autónoma de Nuevo León. Nuevo León, Mexico
| | - Jaime García-Juárez
- Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León. Nuevo León, Mexico
| | - Efrén R Robledo-Leal
- Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León. Nuevo León, México
| | - José G González
- Hospital Universitario, Universidad Autónoma de Nuevo León. Nuevo León, Mexico
| | - Adrian G Rosas-Taraco
- Departamento de Inmunología, Facultad de Medicina, Universidad Autónoma de Nuevo León. Nuevo León, Mexico
| |
Collapse
|
15
|
Rieber N, Singh A, Öz H, Carevic M, Bouzani M, Amich J, Ost M, Ye Z, Ballbach M, Schäfer I, Mezger M, Klimosch SN, Weber ANR, Handgretinger R, Krappmann S, Liese J, Engeholm M, Schüle R, Salih HR, Marodi L, Speckmann C, Grimbacher B, Ruland J, Brown GD, Beilhack A, Loeffler J, Hartl D. Pathogenic fungi regulate immunity by inducing neutrophilic myeloid-derived suppressor cells. Cell Host Microbe 2015; 17:507-14. [PMID: 25771792 PMCID: PMC4400268 DOI: 10.1016/j.chom.2015.02.007] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/17/2014] [Accepted: 01/26/2015] [Indexed: 01/19/2023]
Abstract
Despite continuous contact with fungi, immunocompetent individuals rarely develop pro-inflammatory antifungal immune responses. The underlying tolerogenic mechanisms are incompletely understood. Using both mouse models and human patients, we show that infection with the human pathogenic fungi Aspergillus fumigatus and Candida albicans induces a distinct subset of neutrophilic myeloid-derived suppressor cells (MDSCs), which functionally suppress T and NK cell responses. Mechanistically, pathogenic fungi induce neutrophilic MDSCs through the pattern recognition receptor Dectin-1 and its downstream adaptor protein CARD9. Fungal MDSC induction is further dependent on pathways downstream of Dectin-1 signaling, notably reactive oxygen species (ROS) generation as well as caspase-8 activity and interleukin-1 (IL-1) production. Additionally, exogenous IL-1β induces MDSCs to comparable levels observed during C. albicans infection. Adoptive transfer and survival experiments show that MDSCs are protective during invasive C. albicans infection, but not A. fumigatus infection. These studies define an innate immune mechanism by which pathogenic fungi regulate host defense. Pathogenic fungi induce myeloid-derived suppressor cells (MDSCs) MDSC induction involves Dectin-1/CARD9, ROS, caspase-8, and IL-1 MDSCs dampen T and NK cell immune responses Adoptive transfer of MDSCs improves survival in Candida infection in vivo
Collapse
Affiliation(s)
- Nikolaus Rieber
- Department of Pediatrics I, University of Tübingen, 72076 Tübingen, Germany.
| | - Anurag Singh
- Department of Pediatrics I, University of Tübingen, 72076 Tübingen, Germany
| | - Hasan Öz
- Department of Pediatrics I, University of Tübingen, 72076 Tübingen, Germany
| | - Melanie Carevic
- Department of Pediatrics I, University of Tübingen, 72076 Tübingen, Germany
| | - Maria Bouzani
- Department of Medicine II, University of Würzburg, 97080 Würzburg, Germany
| | - Jorge Amich
- IZKF Research Group for Experimental Stem Cell Transplantation, Department of Medicine II, 97080 Würzburg, Germany
| | - Michael Ost
- Department of Pediatrics I, University of Tübingen, 72076 Tübingen, Germany
| | - Zhiyong Ye
- Department of Pediatrics I, University of Tübingen, 72076 Tübingen, Germany; Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Marlene Ballbach
- Department of Pediatrics I, University of Tübingen, 72076 Tübingen, Germany
| | - Iris Schäfer
- Department of Pediatrics I, University of Tübingen, 72076 Tübingen, Germany
| | - Markus Mezger
- Department of Pediatrics I, University of Tübingen, 72076 Tübingen, Germany
| | - Sascha N Klimosch
- Institute of Cell Biology, Department of Immunology, University of Tübingen, 72076 Tübingen, Germany
| | - Alexander N R Weber
- Institute of Cell Biology, Department of Immunology, University of Tübingen, 72076 Tübingen, Germany
| | | | - Sven Krappmann
- Microbiology Institute - Clinical Microbiology, Immunology and Hygiene, University Hospital of Erlangen and Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Johannes Liese
- Department of Pediatrics, University of Würzburg, 97080 Würzburg, Germany
| | - Maik Engeholm
- Department of Neurology, University of Tübingen, 72076 Tübingen, Germany
| | - Rebecca Schüle
- Department of Neurology, University of Tübingen, 72076 Tübingen, Germany
| | | | - Laszlo Marodi
- Department of Infectious and Pediatric Immunology, Medical and Health Science Center, University of Debrecen, 4032 Debrecen, Hungary
| | - Carsten Speckmann
- Centre of Chronic Immunodeficiency (CCI), University Medical Center Freiburg and University of Freiburg, 79106 Freiburg, Germany
| | - Bodo Grimbacher
- Centre of Chronic Immunodeficiency (CCI), University Medical Center Freiburg and University of Freiburg, 79106 Freiburg, Germany
| | - Jürgen Ruland
- Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Gordon D Brown
- Aberdeen Fungal Group, Section of Immunology and Infection, University of Aberdeen, AB24 3FX Aberdeen, UK
| | - Andreas Beilhack
- IZKF Research Group for Experimental Stem Cell Transplantation, Department of Medicine II, 97080 Würzburg, Germany
| | - Juergen Loeffler
- Department of Medicine II, University of Würzburg, 97080 Würzburg, Germany
| | - Dominik Hartl
- Department of Pediatrics I, University of Tübingen, 72076 Tübingen, Germany.
| |
Collapse
|
16
|
Drummond RA, Wallace C, Reid DM, Way SS, Kaplan DH, Brown GD. Cutting edge: Failure of antigen-specific CD4+ T cell recruitment to the kidney during systemic candidiasis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:5381-5. [PMID: 25344471 PMCID: PMC4238746 DOI: 10.4049/jimmunol.1401675] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 10/09/2014] [Indexed: 11/30/2022]
Abstract
Candida albicans is the leading cause of systemic candidiasis, a fungal disease associated with high mortality and poor treatment options. The kidney is the target organ during infection and whose control is largely dependent on innate immunity, because lymphocytes appear redundant for protection. In this article, we show that this apparent redundancy stems from a failure of Ag-specific CD4(+) T cells to migrate into infected kidneys. In contrast, Ag-specific CD8(+) T cells are recruited normally. Using Ag-loaded immunoliposomes to artificially reverse this defective migration, we show that recruited Ag-specific CD4(+) T cells polarize toward a Th17 phenotype in the kidney and are protective during fungal infection. Therefore, our data explain the redundancy of CD4(+) T cells for defense against systemic infection with C. albicans and have important implications for our understanding of antifungal immunity and the control of renal infections.
Collapse
Affiliation(s)
- Rebecca A Drummond
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, United Kingdom
| | - Carol Wallace
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, United Kingdom
| | - Delyth M Reid
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, United Kingdom
| | - Sing Sing Way
- Division of Infectious Diseases, Cincinnati Children's Hospital, Cincinnati, OH 45229; and
| | - Daniel H Kaplan
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Gordon D Brown
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, United Kingdom;
| |
Collapse
|
17
|
Whibley N, Maccallum DM, Vickers MA, Zafreen S, Waldmann H, Hori S, Gaffen SL, Gow NAR, Barker RN, Hall AM. Expansion of Foxp3(+) T-cell populations by Candida albicans enhances both Th17-cell responses and fungal dissemination after intravenous challenge. Eur J Immunol 2014; 44:1069-83. [PMID: 24435677 PMCID: PMC3992851 DOI: 10.1002/eji.201343604] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 11/15/2013] [Accepted: 01/09/2014] [Indexed: 01/01/2023]
Abstract
Candida albicans remains the fungus most frequently associated with nosocomial bloodstream infection. In disseminated candidiasis, the role of Foxp3(+) regulatory T (Treg) cells remains largely unexplored. Our aims were to characterize Foxp3(+) Treg-cell activation in a murine intravenous challenge model of disseminated C. albicans infection, and determine the contribution to disease. Flow cytometric analyses demonstrated that C. albicans infection drove in vivo expansion of a splenic CD4(+) Foxp3(+) population that correlated positively with fungal burden. Depletion from Foxp3(hCD2) reporter mice in vivo confirmed that Foxp3(+) cells exacerbated fungal burden and inflammatory renal disease. The CD4(+) Foxp3(+) population expanded further after in vitro stimulation with C. albicans antigens (Ags), and included at least three cell types. These arose from proliferation of the natural Treg-cell subset, together with conversion of Foxp3(-) cells to the induced Treg-cell form, and to a cell type sharing effector Th17-cell characteristics, expressing ROR-γt, and secreting IL-17A. The expanded Foxp3(+) T cells inhibited Th1 and Th2 responses, but enhanced Th17-cell responses to C. albicans Ags in vitro, and in vivo depletion confirmed their ability to enhance the Th17-cell response. These data lead to a model for disseminated candidiasis whereby expansion of Foxp3(+) T cells promotes Th17-cell responses that drive pathology.
Collapse
Affiliation(s)
- Natasha Whibley
- Division of Applied Medicine, University of Aberdeen, Aberdeen, UK; Aberdeen Fungal Group, University of Aberdeen, Aberdeen, UK; Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
The mouse intravenous (IV) challenge model of Candida albicans invasive fungal infection has been widely used to study the importance of the innate immune system in these infections. This chapter describes this well-characterized model, where fungal cells are administered directly into the mouse bloodstream to initiate a systemic infection. The preparation of tissue samples from infected mice to allow evaluation of disease progression and host responses is also described.
Collapse
|
19
|
Huang PY, Hung MH, Shie SS, Su LH, Chen KY, Ye JJ, Chiang PC, Leu HS, Huang CT. Molecular concordance of concurrent Candida albicans candidemia and candiduria. Diagn Microbiol Infect Dis 2013; 76:382-4. [DOI: 10.1016/j.diagmicrobio.2013.03.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2012] [Revised: 03/13/2013] [Accepted: 03/13/2013] [Indexed: 10/26/2022]
|
20
|
Canadian clinical practice guidelines for invasive candidiasis in adults. CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY 2012; 21:e122-50. [PMID: 22132006 DOI: 10.1155/2010/357076] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Candidemia and invasive candidiasis (C/IC) are life-threatening opportunistic infections that add excess morbidity, mortality and cost to the management of patients with a range of potentially curable underlying conditions. The Association of Medical Microbiology and Infectious Disease Canada developed evidence-based guidelines for the approach to the diagnosis and management of these infections in the ever-increasing population of at-risk adult patients in the health care system. Over the past few years, a new and broader understanding of the epidemiology and pathogenesis of C/IC has emerged and has been coupled with the availability of new antifungal agents and defined strategies for targeting groups at risk including, but not limited to, acute leukemia patients, hematopoietic stem cell transplants and solid organ transplants, and critical care unit patients. Accordingly, these guidelines have focused on patients at risk for C/IC, and on approaches of prevention, early therapy for suspected but unproven infection, and targeted therapy for probable and proven infection.
Collapse
|
21
|
Localized candidiasis in kidney presented as a mass mimicking renal cell carcinoma. Case Rep Infect Dis 2012; 2012:953590. [PMID: 22567490 PMCID: PMC3336244 DOI: 10.1155/2012/953590] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 01/30/2012] [Indexed: 12/16/2022] Open
Abstract
Candida albicans is a ubiquitous fungus and infection of urinary tract by C. albicans can be originated from blood or retrograde infection. We reported a case of localized candidiasis in the kidney presenting as a mass. The patient was a 61-year-old male with a history of type 2 diabetes mellitus and urinary bladder urothelial carcinoma status post radical cystoprostatectomy with a neobladder three years ago. Pathology at that time also showed a prostatic adenocarcinoma (Gleason score 3 + 4) in addition to the high-grade urothelial carcinoma. Three month ago the patient presented with flank pain, chill, and increased white cell counts. Imaging study showed a large renal mass suspicious for a renal cell carcinoma. Radical nephrectomy was performed and found that there was a large pocket of pus in the retroperitoneum around the right kidney during the surgery. Intraoperative abscess cultures were positive for C. albicans. Pathology showed a 13.5 cm necrotic renal mass extending to the perinephric fat. Histologically the tumor showed necrotic granulomatous inflammation. Grocott stain in the surgical specimen was positive for pseudohyphae and yeast forms. The patient was initiated a course of fluconazole postoperatively and was feeling well.
Collapse
|
22
|
Lin L, Ibrahim AS, Baquir B, Palosaari A, Spellberg B. Luminescent-activated transfected killer cells to monitor leukocyte trafficking during systemic bacterial and fungal infection. J Infect Dis 2011; 205:337-47. [PMID: 22124127 DOI: 10.1093/infdis/jir725] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Activated transfected killer (ATAK) cells are immortal phagocytes transfected with a luminescence reporter that effectively treat lethal infections in neutropenic mice. Their in vivo trafficking, lifespan, and immunogenicity are unknown. METHODS Mice were made neutropenic; infected or not with Staphylococcus aureus, Acinetobacter baumannii, Candida albicans, or Aspergillus fumigatus; and treated intraperitoneally with ATAK cells. Cell trafficking and lifespan were assessed by in vivo imaging and reverse transcription-polymerase chain reaction. RESULTS In uninfected neutropenic mice, ATAK cells spread from the mesentery into visceral organs on days 1-3. Splenic accumulation of ATAK cells increased at day 1 after infection with S. aureus and A. baumannii, and kidney accumulation increased in mice infected with C. albicans. Lung accumulation was seen at day 3 in mice infected by inhalation with A. fumigatus. By day 8, coincident with increasing anti-ATAK antibodies, luminescence signal was lost and there was no detectable mRNA transcription from ATAK cells. CONCLUSIONS ATAK cells accumulated in target organs with distinct profiles, depending on the microbial etiology of infection. Finally, generation of an anti-ATAK immune response may provide an important safety mechanism that helps clear the cells from the host as the marrow recovers.
Collapse
Affiliation(s)
- Lin Lin
- Department of Medicine, Division of General Internal Medicine, Los Angeles Biomedical Research Institute, Harbor-University of California at Los Angeles Medical Center, Torrance, CA, USA
| | | | | | | | | |
Collapse
|
23
|
Chaturvedi AK, Lazzell AL, Saville SP, Wormley FL, Monteagudo C, Lopez-Ribot JL. Validation of the tetracycline regulatable gene expression system for the study of the pathogenesis of infectious disease. PLoS One 2011; 6:e20449. [PMID: 21633704 PMCID: PMC3102114 DOI: 10.1371/journal.pone.0020449] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Accepted: 04/20/2011] [Indexed: 12/28/2022] Open
Abstract
Understanding the pathogenesis of infectious disease requires the examination and successful integration of parameters related to both microbial virulence and host responses. As a practical and powerful method to control microbial gene expression, including in vivo, the tetracycline-regulatable system has recently gained the favor of many investigative groups. However, some immunomodulatory effects of the tetracyclines, including doxycycline, could potentially limit its use to evaluate host responses during infection. Here we have used a well-established murine model of disseminated candidiasis, which is highly dependent on both the virulence displayed by the fungal cells and on the host immune status, to validate the use of this system. We demonstrate that the pathogenesis of the wild type C. albicans CAF2-1 strain, which does not contain any tet-regulatable element, is not affected by the presence of doxycycline. Moreover levels of key cytokines, chemokines and many other biomarkers, as determined by multi-analyte profiling, remain essentially unaltered by the presence of the antibiotic during infection. Our results indicate that the levels of doxycycline needed to control the tetracycline regulatable promoter gene expression system have no detectable effect on global host responses during candidiasis. Because tet-regulatable systems are now being increasingly used in a variety of pathogenic microorganisms, these observations have wide implications in the field of infectious diseases.
Collapse
Affiliation(s)
- Ashok K. Chaturvedi
- Department of Biology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, Texas, United States of America
| | - Anna L. Lazzell
- Department of Biology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, Texas, United States of America
| | - Stephen P. Saville
- Department of Biology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, Texas, United States of America
| | - Floyd L. Wormley
- Department of Biology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, Texas, United States of America
| | - Carlos Monteagudo
- Departmento de Patología, Facultad de Medicina y Odontología, Universidad de Valencia, Valencia, Spain
| | - Jose L. Lopez-Ribot
- Department of Biology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|
24
|
Candida and invasive candidiasis: back to basics. Eur J Clin Microbiol Infect Dis 2011; 31:21-31. [PMID: 21544694 DOI: 10.1007/s10096-011-1273-3] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2010] [Accepted: 04/12/2011] [Indexed: 10/18/2022]
Abstract
The ubiquitous Candida spp. is an opportunistic fungal pathogen which, despite treatment with antifungal drugs, can cause fatal bloodstream infections (BSIs) in immunocompromised and immunodeficient persons. Thus far, several major C. albicans virulence factors have been relatively well studied, including morphology switching and secreted degradative enzymes. However, the exact mechanism of Candida pathogenesis and the host response to invasion are still not well elucidated. The relatively recent discovery of the quorum-sensing molecule farnesol and the existence of quorum sensing as a basic regulatory phenomenon of the C. albicans population behavior has revolutionized Candida research. Through population density regulation, the quorum-sensing mechanism also controls the cellular morphology of a C. albicans population in response to environmental factors, thereby, effectively placing morphology switching downstream of quorum sensing. Thus, the quorum-sensing phenomenon has been hailed as the 'missing piece' of the pathogenicity puzzle. Here, we review what is known about Candida spp. as the etiological agents of invasive candidiasis and address our current understanding of the quorum-sensing phenomenon in relation to virulence in the host.
Collapse
|
25
|
Fisher JF, Kavanagh K, Sobel JD, Kauffman CA, Newman CA. Candida Urinary Tract Infection: Pathogenesis. Clin Infect Dis 2011; 52 Suppl 6:S437-51. [DOI: 10.1093/cid/cir110] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
26
|
Cleary IA, Reinhard SM, Miller CL, Murdoch C, Thornhill MH, Lazzell AL, Monteagudo C, Thomas DP, Saville SP. Candida albicans adhesin Als3p is dispensable for virulence in the mouse model of disseminated candidiasis. MICROBIOLOGY-SGM 2011; 157:1806-1815. [PMID: 21436220 DOI: 10.1099/mic.0.046326-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The presence of specific proteins, including Ece1p, Hwp1p and Als3p, distinguishes the Candida albicans hyphal cell wall from that of yeast-form cells. These proteins are thought to be important for the ability of C. albicans cells to adhere to living and non-living surfaces and for the cell-to-cell adhesion necessary for biofilm formation, and also to be pivotal in mediating C. albicans interactions with endothelial cells. Using an in vitro flow adhesion assay, we previously observed that yeast cells bind in greater numbers to human microvascular endothelial cells than do hyphal or pseudohyphal cells. This is consistent with previous observations that, in a murine model of disseminated candidiasis, cells locked in the yeast form can efficiently escape the bloodstream and invade host tissues. To more precisely explore the role of Als3p in adhesion and virulence, we deleted both copies of ALS3 in a wild-type C. albicans strain. In agreement with previous studies, our als3Δ null strain formed hyphae normally but was defective in biofilm formation. Whilst ALS3 was not expressed in our null strain, hypha-specific genes such as ECE1 and HWP1 were still induced appropriately. Both the yeast form and the hyphal form of the als3Δ strain adhered to microvascular endothelial cells to the same extent as a wild-type strain under conditions of flow, indicating that Als3p is not a significant mediator of the initial interaction between fungal cells and the endothelium. Finally, in a murine model of haematogenously disseminated candidiasis the mutant als3Δ remained as virulent as the wild-type parent strain.
Collapse
Affiliation(s)
- Ian A Cleary
- Department of Biology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA
| | - Sara M Reinhard
- Department of Biology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA
| | - C Lindsay Miller
- Department of Biology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA
| | - Craig Murdoch
- Academic Unit of Oral and Maxillofacial Medicine and Surgery, University of Sheffield, Sheffield, UK
| | - Martin H Thornhill
- Academic Unit of Oral and Maxillofacial Medicine and Surgery, University of Sheffield, Sheffield, UK
| | - Anna L Lazzell
- Department of Biology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA
| | - Carlos Monteagudo
- Departmento de Patología, Facultad de Medicina y Odontología, Universidad de Valencia, 46010 Valencia, Spain
| | - Derek P Thomas
- Department of Biology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA
| | - Stephen P Saville
- Department of Biology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA
| |
Collapse
|
27
|
Szabo EK, MacCallum DM. The contribution of mouse models to our understanding of systemic candidiasis. FEMS Microbiol Lett 2011; 320:1-8. [PMID: 21395661 DOI: 10.1111/j.1574-6968.2011.02262.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Some Candida species are common commensals, which can become opportunistic pathogens in susceptible hosts. In severely ill patients, Candida species, particularly Candida albicans, can cause life-threatening systemic infections. These infections are difficult to diagnose, as symptoms are similar to those of systemic bacterial infections. These difficulties can lead to delays in initiation in antifungal therapy, which contributes to the high mortality rates (> 40%) associated with these infections. In order to investigate systemic Candida infection, mouse models have been developed that mimic human disease, the most common being the intravenous infection model and the gastrointestinal colonization and dissemination model. This review discusses the two models and the contributions that they have made to our understanding of fungal virulence, host response to infection and the development of novel antifungal therapies and diagnostics.
Collapse
Affiliation(s)
- Edina K Szabo
- Aberdeen Fungal Group, School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Foresterhill, UK
| | | |
Collapse
|
28
|
Lim CSY, Rosli R, Seow HF, Chong PP. Transcriptome profiling of endothelial cells during infections with high and low densities of C. albicans cells. Int J Med Microbiol 2011; 301:536-46. [PMID: 21371935 DOI: 10.1016/j.ijmm.2010.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Revised: 12/06/2010] [Accepted: 12/07/2010] [Indexed: 01/04/2023] Open
Abstract
Systemic infections of Candida albicans, the most prevalent fungal pathogen in humans, are on the rise in recent years. However, the exact mode of pathogenesis of this fungus is still not well elucidated. Previous studies using C. albicans mutants locked into the yeast form via gene deletion found that this form was avirulent and did not induce significant differential expression of host genes in vitro. In this study, a high density of C. albicans was used to infect human umbilical vein endothelial cells (HUVEC), resulting in yeast-form infections, whilst a low density of C. albicans resulted in hyphae infections. Transcriptional profiling of HUVEC response to these infections showed that high densities of C. albicans induced a stronger, broader transcriptional response from HUVEC than low densities of C. albicans infection. Many of the genes that were significantly differentially expressed were involved in apoptosis and cell death. In addition, conditioned media from the high-density infections caused a significant reduction in HUVEC viability, suggesting that certain molecules released during C. albicans and HUVEC interactions were capable of causing cell death. This study has shown that C. albicans yeast-forms, at high densities, cannot be dismissed as avirulent, but instead could possibly contribute to C. albicans pathogenesis.
Collapse
Affiliation(s)
- Crystale S Y Lim
- Dept. of Biomedical Sciences, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | | | | | | |
Collapse
|
29
|
Nelson MP, Christmann BS, Werner JL, Metz AE, Trevor JL, Lowell CA, Steele C. IL-33 and M2a alveolar macrophages promote lung defense against the atypical fungal pathogen Pneumocystis murina. THE JOURNAL OF IMMUNOLOGY 2011; 186:2372-81. [PMID: 21220696 DOI: 10.4049/jimmunol.1002558] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We have recently reported that mice deficient in the myeloid Src-family tyrosine kinases Hck, Fgr, and Lyn (Src triple knockout [TKO]) had augmented innate lung clearance of Pneumocystis murina that correlated with a higher ability of alveolar macrophages (AMs) from these mice to kill P. murina. In this article, we show that despite possessing enhanced killing, AMs from naive Src TKO mice did not demonstrate enhanced inflammatory responses to P. murina. We subsequently discovered that both AMs and lungs from P. murina-infected Src TKO mice expressed significantly greater levels of the M2a markers RELM-α and Arg1, and the M2a-associated chemokines CCL17 and CCL22 than did wild-type mice. IL-4 and IL-13, the primary cytokines that promote M2a polarization, were not differentially produced in the lungs between wild-type and Src TKO mice. P. murina infection in Src TKO mice resulted in enhanced lung production of the novel IL-1 family cytokine IL-33. Immunohistochemical analysis of IL-33 in lung tissue revealed localization predominantly in the nucleus of alveolar epithelial cells. We further demonstrate that experimental polarization of naive AMs to M2a resulted in more efficient killing of P. murina compared with untreated AMs, which was further enhanced by the addition of IL-33. Administration of IL-33 to C57BL/6 mice increased lung RELM-α and CCL17 levels, and enhanced clearance of P. murina, despite having no effect on the cellular composition of the lungs. Collectively, these results indicate that M2a AMs are potent effector cells against P. murina. Furthermore, enhancing M2a polarization may be an adjunctive therapy for the treatment of Pneumocystis.
Collapse
Affiliation(s)
- Michael P Nelson
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Kundu G, Noverr MC. Exposure to host or fungal PGE₂ abrogates protection following immunization with Candida-pulsed dendritic cells. Med Mycol 2010; 49:380-94. [PMID: 21077736 DOI: 10.3109/13693786.2010.532514] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Candida albicans produces an immunomodulatory oxylipin from arachidonic acid that is structurally identical to host prostaglandin E₂ (PGE₂). In terms of host immune responses, PGE₂ can promote Th2 responses, which are non-protective against fungal infections. We investigated the effect of host or fungal PGE₂ on murine bone marrow-derived dendritic cell (DC) cytokine production, and the ability to immunize mice against systemic infection with C. albicans. We used GM-CSF to produce myeloid DCs (GM-DCs) and FLT-3L to enrich for plasmacytoid DCs (FL-DCs). In the presence of hyphae, PGE₂ promoted Th2 cytokine production and suppressed Th1 cytokine production. Immunization with yeast-pulsed DCs but not hyphae-pulsed DCs lead to a reduction in kidney fungal burden during systemic infection, which was most dramatic with FL-DCs. However, exposure to either host or fungal PGE₂ during antigenic stimulation abrogated the ability of yeast-pulsed DCs to protect against infection. The lack of protection was associated with a trend towards reduced Th1 cytokines and increased Th2 cytokines in the spleen. However, the pattern of protection did not completely match cytokine expression. Locally, in FL-DC pulsed mice, reduced Th1 and exacerbated Th2 and Th17 cytokines were only detected in the kidneys of mice that did not show reductions in fungal burden after vaccination. This indicates that host or fungal PGE₂ can shift adaptive responses in favor of the pathogen and that uncontrolled Th17 responses are detrimental during systemic infection.
Collapse
Affiliation(s)
- Gitanjali Kundu
- Department of Immunology & Microbiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | | |
Collapse
|
31
|
Limited role of secreted aspartyl proteinases Sap1 to Sap6 in Candida albicans virulence and host immune response in murine hematogenously disseminated candidiasis. Infect Immun 2010; 78:4839-49. [PMID: 20679440 DOI: 10.1128/iai.00248-10] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Candida albicans secreted aspartyl proteinases (Saps) are considered virulence-associated factors. Several members of the Sap family were claimed to play a significant role in the progression of candidiasis established by the hematogenous route. This assumption was based on the observed attenuated virulence of sap-null mutant strains. However, the exclusive contribution of SAP genes to their attenuated phenotype was not unequivocally confirmed, as the Ura status of these mutant strains could also have contributed to the attenuation. In this study, we have reassessed the importance of SAP1 to SAP6 in a murine model of hematogenously disseminated candidiasis using sap-null mutant strains not affected in their URA3 gene expression and compared their virulence phenotypes with those of Ura-blaster sap mutants. The median survival time of BALB/c mice intravenously infected with a mutant strain lacking SAP1 to SAP3 was equivalent to that of mice infected with wild-type strain SC5314, while those infected with mutant strains lacking SAP5 showed slightly extended survival times. Nevertheless, no differences could be observed between the wild type and a Δsap456 mutant in their abilities to invade mouse kidneys. Likewise, a deficiency in SAP4 to SAP6 had no noticeable impact on the immune response elicited in the spleens and kidneys of C. albicans-infected mice. These results contrast with the behavior of equivalent Ura-blaster mutants, which presented a significant reduction in virulence. Our results suggest that Sap1 to Sap6 do not play a significant role in C. albicans virulence in a murine model of hematogenously disseminated candidiasis and that, in this model, Sap1 to Sap3 are not necessary for successful C. albicans infection.
Collapse
|
32
|
Lin L, Ibrahim AS, Baquir B, Fu Y, Applebaum D, Schwartz J, Wang A, Avanesian V, Spellberg B. Safety and efficacy of activated transfected killer cells for neutropenic fungal infections. J Infect Dis 2010; 201:1708-17. [PMID: 20397927 DOI: 10.1086/652496] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Invasive fungal infections cause considerable morbidity and mortality in neutropenic patients. White blood cell transfusions are a promising treatment for such infections, but technical barriers have prevented their widespread use. METHODS To recapitulate white blood cell transfusions, we are developing a cell-based immunotherapy using a phagocytic cell line, HL-60. We sought to stably transfect HL-60 cells with a suicide trap (herpes simplex virus thymidine kinase), to enable purging of the cells when desired, and a bioluminescence marker, to track the cells in vivo in mice. RESULTS Transfection was stable despite 20 months of continuous culture or storage in liquid nitrogen. Activation of these transfected cells with retinoic acid and dimethyl sulfamethoxazole enhanced their microbicidal effects. Activated transfected killer (ATAK) cells were completely eliminated after exposure to ganciclovir, confirming function of the suicide trap. ATAK cells improved the survival of neutropenic mice with lethal disseminated candidiasis and inhalational aspergillosis. Bioluminescence and histopathologic analysis confirmed that the cells were purged from surviving mice after ganciclovir treatment. Comprehensive necropsy, histopathology, and metabolomic analysis revealed no toxicity of the cells. CONCLUSIONS These results lay the groundwork for continued translational development of this promising, novel technology for the treatment of refractory infections in neutropenic hosts.
Collapse
Affiliation(s)
- Lin Lin
- Division of Infectious Diseases, Los Angeles Biomedical Research Institute, Harbor-University of California at Los Angeles (UCLA) Medical Center, 1124 West Carson Street, Torrance, CA 90502, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Lin L, Ibrahim AS, Xu X, Farber JM, Avanesian V, Baquir B, Fu Y, French SW, Edwards JE, Spellberg B. Th1-Th17 cells mediate protective adaptive immunity against Staphylococcus aureus and Candida albicans infection in mice. PLoS Pathog 2009; 5:e1000703. [PMID: 20041174 PMCID: PMC2792038 DOI: 10.1371/journal.ppat.1000703] [Citation(s) in RCA: 369] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2009] [Accepted: 11/19/2009] [Indexed: 01/17/2023] Open
Abstract
We sought to define protective mechanisms of immunity to Staphylococcus aureus and Candida albicans bloodstream infections in mice immunized with the recombinant N-terminus of Als3p (rAls3p-N) vaccine plus aluminum hydroxide (Al(OH3) adjuvant, or adjuvant controls. Deficiency of IFN-γ but not IL-17A enhanced susceptibility of control mice to both infections. However, vaccine-induced protective immunity against both infections required CD4+ T-cell-derived IFN-γ and IL-17A, and functional phagocytic effectors. Vaccination primed Th1, Th17, and Th1/17 lymphocytes, which produced pro-inflammatory cytokines that enhanced phagocytic killing of both organisms. Vaccinated, infected mice had increased IFN-γ, IL-17, and KC, increased neutrophil influx, and decreased organism burden in tissues. In summary, rAls3p-N vaccination induced a Th1/Th17 response, resulting in recruitment and activation of phagocytes at sites of infection, and more effective clearance of S. aureus and C. albicans from tissues. Thus, vaccine-mediated adaptive immunity can protect against both infections by targeting microbes for destruction by innate effectors. The bacterium Staphylococcus aureus and the fungus Candida are the second and third leading cause of bloodstream infections in hospitalized patients. A vaccine to prevent such infections would be of enormous public health benefit. The leading hypothesis to explain why vaccines have not been successfully developed against these infections is that the microbes causing the infections are highly complex, and use multiple weapons (so-called “virulence factors”) to cause disease in humans. Therefore, a vaccine targeting either infection would have to neutralize many of these virulence factors at the same time. We have been developing a vaccine that simultaneously targets both types of infections. Our vaccine is based on a single virulence factor used by Candida, which has a similar shape to virulence factors used by S. aureus. In the current study, we report that our vaccine induces specialized cells in the immune system to more effectively call in reinforcements to kill the organisms. These data demonstrate that vaccines against both organisms can be developed even if they do not work by neutralizing multiple virulence factors, and therefore open the door to a far wider array of vaccine types against both infections.
Collapse
Affiliation(s)
- Lin Lin
- The Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
| | - Ashraf S. Ibrahim
- The Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
- The David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Xin Xu
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Joshua M. Farber
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Valentina Avanesian
- The Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
| | - Beverlie Baquir
- The Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
| | - Yue Fu
- The Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
- The David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Samuel W. French
- The David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- The Department of Pathology, Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - John E. Edwards
- The Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
- The David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Brad Spellberg
- The Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-University of California at Los Angeles (UCLA) Medical Center, Torrance, California, United States of America
- The David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- The Division of General Internal Medicine, Harbor-UCLA Medical Center, Torrance, California, United States of America
- * E-mail:
| |
Collapse
|
34
|
MacCallum DM. Massive induction of innate immune response to Candida albicans in the kidney in a murine intravenous challenge model. FEMS Yeast Res 2009; 9:1111-22. [PMID: 19845042 PMCID: PMC2784217 DOI: 10.1111/j.1567-1364.2009.00576.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
In the experimental Candida albicans intravenous challenge model, the kidney is one of the main organs involved in disease. In progressive infection, fungal burdens are found to increase over time, with rapid increases occurring from 24 h postinfection. Renal transcriptional responses were analyzed at this time in the kidneys of mice infected by either a virulent or an attenuated C. albicans strain, allowing comparison of host responses in progressive and nonprogressive infection. The results of this study demonstrate that both infections share a common transcriptional response, consisting of functions associated with the acute-phase reaction. In addition, challenge with the virulent strain led to a massively increased expression of cytokine genes, other innate response genes and genes suggestive of initiation of the adaptive immune response. This immune response to C. albicans infection, which occurs only in progressive infection, may contribute to development of sepsis and, ultimately, host death.
Collapse
Affiliation(s)
- Donna M MacCallum
- Aberdeen Fungal Group, School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2D, UK.
| |
Collapse
|
35
|
MacCallum DM, Castillo L, Brown AJP, Gow NAR, Odds FC. Early-expressed chemokines predict kidney immunopathology in experimental disseminated Candida albicans infections. PLoS One 2009; 4:e6420. [PMID: 19641609 PMCID: PMC2712765 DOI: 10.1371/journal.pone.0006420] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2009] [Accepted: 06/26/2009] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The mouse intravenous challenge model of Candida albicans infection is widely used to determine aspects of host-fungus interaction. We investigated the production of cytokines in the kidneys and spleen of animals up to 48 h after challenge with virulent and attenuated isolates and related these responses to semi-quantitative estimations of histopathological changes in the kidney. METHODOLOGY/PRINCIPAL FINDINGS Progression of Candida albicans infection of the kidney in response to highly virulent fungal strains was characterized by higher levels of host cellular infiltrate, higher lesion densities and greater quantities of fungal elements at 24 and 48 h, and by higher kidney concentrations of IL-1beta, MCP-1, KC, IL-6, G-CSF, TNF, MIP-2 and MIP-1beta, among the immune effectors measured. Levels of the chemokine KC as early as 12 h after challenge correlated significantly with all later measurements of lesion severity. Early renal IL-6 and MIP-1beta concentrations also correlated with subsequent damage levels, but less significantly than for KC. All chemokines tested appeared in kidney homogenates, while most of the cytokines were undetectable in kidney and spleen homogenates. GM-CSF and IL-10 showed inverse correlations with measures of lesion severity, suggesting these alone may have exerted a defensive role. Spleen levels of KC at all times showed significant associations with kidney lesion measurements. CONCLUSIONS/SIGNIFICANCE Elevated chemokine levels, including KC, represent the earliest responses to C. albicans infection in the mouse kidney. Fungal strains of low mouse virulence stimulate a lower innate response and less host infiltrate than more virulent strains. These findings are consistent with immunopathological damage to kidneys in the mouse C. albicans infection model and with growing evidence implicating some TLR pathways as the main point of interaction between fungal surface polysaccharides and leukocytes.
Collapse
Affiliation(s)
- Donna M. MacCallum
- Aberdeen Fungal Group, Institute of Medical Sciences, Aberdeen, United Kingdom
| | - Luis Castillo
- Aberdeen Fungal Group, Institute of Medical Sciences, Aberdeen, United Kingdom
| | | | - Neil A. R. Gow
- Aberdeen Fungal Group, Institute of Medical Sciences, Aberdeen, United Kingdom
| | - Frank C. Odds
- Aberdeen Fungal Group, Institute of Medical Sciences, Aberdeen, United Kingdom
- * E-mail:
| |
Collapse
|
36
|
Adhesion of Candida albicans to endothelial cells under physiological conditions of flow. Infect Immun 2009; 77:3872-8. [PMID: 19581400 DOI: 10.1128/iai.00518-09] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Candida albicans is a commensal organism that under certain circumstances can become pathogenic. During systemic infection C. albicans is disseminated via the circulation to distant organs, where it causes multiple organ failure. Despite the severity of systemic C. albicans infection, little is known about the mechanisms involved in the adhesion of this organism to the endothelium lining blood vessels. Previous studies have used static assays to examine adhesion. However, these do not realistically model blood vessels, where circulating C. albicans cells must adhere to the endothelium under conditions of flow and shear stress. Furthermore, there is conflicting evidence concerning the role played by yeast, pseudohyphal, and hyphal forms of C. albicans in adhesion to endothelium. To test the hypothesis that there may be differences in the abilities of these three morphogenic forms of C. albicans to adhere to endothelium under conditions of flow, we developed an in vitro flow adhesion assay. We found that all three forms of C. albicans rapidly bound to confluent endothelial cells under conditions of flow. Maximum adhesion was found at low shear stress levels similar to that found in postcapillary venules. Moreover, yeast forms bound in significantly greater numbers than did pseudohyphal and hyphal forms, respectively, contrasting with previous findings from static assays. These findings are consistent with recent in vivo data suggesting that yeast forms may be capable of adhering to the endothelium and migrating into the tissues before undergoing morphogenic change to cause tissue damage.
Collapse
|
37
|
The antifungal vaccine derived from the recombinant N terminus of Als3p protects mice against the bacterium Staphylococcus aureus. Infect Immun 2008; 76:4574-80. [PMID: 18644876 DOI: 10.1128/iai.00700-08] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Vaccination with the recombinant N terminus of the candidal adhesin Als3p (rAls3p-N) protects mice from lethal candidemia. Candidal Als3p also is structurally similar to the microbial surface components recognizing adhesive matrix molecule adhesin, clumping factor, from Staphylococcus aureus. To determine the potential for cross-kingdom vaccination, we immunized mice with rAls3p-N or negative control proteins and challenged them via the tail vein with S. aureus or other gram-positive or gram-negative pathogens. The rAls3p-N vaccine, but neither tetanus toxoid nor a related Als protein (Als5p), improved the survival of vaccinated mice subsequently infected with multiple clinical isolates of S. aureus, including methicillin-resistant strains. The rAls3p-N vaccine was effective against S. aureus when combined with aluminum hydroxide adjuvant. However, the vaccine did not improve the survival of mice infected with other bacterial pathogens. Vaccinated, infected mice mounted moderated type 1 immune responses. T lymphocyte-deficient mice were more susceptible to S. aureus infection, but B lymphocyte-deficient mice were not. Furthermore, T but not B lymphocytes from vaccinated mice mediated protection in adoptive transfer studies. The passive transfer of immune serum was not protective. These data provide the foundation for cross-kingdom vaccine development against S. aureus and Candida, which collectively cause 200,000 bloodstream infections resulting in >/=40,000 to 50,000 deaths annually in the United States alone.
Collapse
|
38
|
Barker K, Park H, Phan Q, Xu L, Homayouni R, Rogers P, Filler S. Transcriptome Profile of the Vascular Endothelial Cell Response toCandida albicans. J Infect Dis 2008; 198:193-202. [DOI: 10.1086/589516] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
39
|
Spellberg B. Novel insights into disseminated candidiasis: pathogenesis research and clinical experience converge. PLoS Pathog 2008; 4:e38. [PMID: 18282100 PMCID: PMC2242839 DOI: 10.1371/journal.ppat.0040038] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Brad Spellberg
- Division of Infectious Diseases, Harbor-University of California Los Angeles Medical Center, Torrance, California, United States of America.
| |
Collapse
|
40
|
Spellberg B, Ibrahim AS, Lin L, Avanesian V, Fu Y, Lipke P, Otoo H, Ho T, Edwards JE. Antibody titer threshold predicts anti-candidal vaccine efficacy even though the mechanism of protection is induction of cell-mediated immunity. J Infect Dis 2008; 197:967-71. [PMID: 18419471 DOI: 10.1086/529204] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
We previously reported that vaccination with Freund's adjuvant plus the recombinant N-terminus of the candidal adhesin, Als3p (rAls3p-N), protects mice from disseminated candidiasis. Here we report that the rAls3p-N vaccine is effective when combined with aluminum hydroxide adjuvant. Antibody titers of > or =1:6400 accurately predicted protection from infection. Nevertheless, neither B lymphocytes nor serum from immunized animals transferred protection to vaccine-naive animals. In contrast, CD3(+), CD4(+), or CD8(+) T lymphocytes from immunized animals transferred protection, and the vaccine was efficacious in IL-4-deficient mice but not in IFN-gamma-deficient mice. These data have significant implications for the development and interpretation of vaccine surrogate markers.
Collapse
Affiliation(s)
- Brad Spellberg
- Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-University of California, Los Angeles, Medical Center, Torrance, CA 90502, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Considerable differences in vaccine immunogenicities and efficacies related to the diluent used for aluminum hydroxide adjuvant. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2008; 15:582-4. [PMID: 18184821 DOI: 10.1128/cvi.00427-07] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We are developing an anticandidal vaccine using the recombinant N terminus of Als3p (rAls3p-N). We report that although more rAls3p-N was bound by aluminum hydroxide diluted in saline than by aluminum hydroxide diluted in phosphate-buffered saline (PBS), its immunogenicity and efficacy were superior in PBS. Thus, protein binding, by itself, may not predict the efficacy of some vaccines with aluminum adjuvants.
Collapse
|
42
|
Ibrahim AS, Gebermariam T, Fu Y, Lin L, Husseiny MI, French SW, Schwartz J, Skory CD, Edwards JE, Spellberg BJ. The iron chelator deferasirox protects mice from mucormycosis through iron starvation. J Clin Invest 2007; 117:2649-57. [PMID: 17786247 PMCID: PMC1957543 DOI: 10.1172/jci32338] [Citation(s) in RCA: 229] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2007] [Accepted: 06/13/2007] [Indexed: 01/19/2023] Open
Abstract
Mucormycosis causes mortality in at least 50% of cases despite current first-line therapies. Clinical and animal data indicate that the presence of elevated available serum iron predisposes the host to mucormycosis. Here we demonstrate that deferasirox, an iron chelator recently approved for use in humans by the US FDA, is a highly effective treatment for mucormycosis. Deferasirox effectively chelated iron from Rhizopus oryzae and demonstrated cidal activity in vitro against 28 of 29 clinical isolates of Mucorales at concentrations well below clinically achievable serum levels. When administered to diabetic ketoacidotic or neutropenic mice with mucormycosis, deferasirox significantly improved survival and decreased tissue fungal burden, with an efficacy similar to that of liposomal amphotericin B. Deferasirox treatment also enhanced the host inflammatory response to mucormycosis. Most importantly, deferasirox synergistically improved survival and reduced tissue fungal burden when combined with liposomal amphotericin B. These data support clinical investigation of adjunctive deferasirox therapy to improve the poor outcomes of mucormycosis with current therapy. As iron availability is integral to the pathogenesis of other infections (e.g., tuberculosis, malaria), broader investigation of deferasirox as an antiinfective treatment is warranted.
Collapse
Affiliation(s)
- Ashraf S Ibrahim
- Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California 90502, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Navarathna DHMLP, Nickerson KW, Duhamel GE, Jerrels TR, Petro TM. Exogenous farnesol interferes with the normal progression of cytokine expression during candidiasis in a mouse model. Infect Immun 2007; 75:4006-11. [PMID: 17517874 PMCID: PMC1951970 DOI: 10.1128/iai.00397-07] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Candida albicans, a dimorphic fungus composed of yeast and mycelial forms, is the most common human fungal pathogen. Th1 cytokines such as interleukin-2 (IL-2), gamma interferon (IFN-gamma), and tumor necrosis factor alpha (TNF-alpha), which are induced by macrophage IL-12, are critical to resistance against systemic candidiasis, while Th2 cytokines such as IL-4 and IL-5 are less critical. Farnesol is a quorum-sensing molecule produced by C. albicans that controls the formation of mycelia but is also a virulence factor. To determine whether farnesol enhances the virulence of C. albicans by modulating the production of Th1 and Th2 cytokines, mice were pretreated with farnesol prior to intravenous infection with a sublethal dose of farnesol-producing C. albicans. Production of IL-2, IL-4, IL-5, TNF-alpha, IFN-gamma, and IL-12 was evaluated by bead-array flow cytometry and enzyme-linked immunosorbent assay. Mice exhibited an elevation in serum TNF-alpha levels at 48 h and an elevation in IFN-gamma and IL-12 levels at 6 to 12 h after infection with C. albicans. Pretreatment with farnesol significantly reduced the elevation of both IFN-gamma and IL-12 but not TNF-alpha. In contrast, mice pretreated with farnesol exhibited an unexpected elevation in IL-5 levels. To determine whether farnesol has a direct effect on macrophage production of IL-12, peritoneal macrophages were pretreated with farnesol prior to stimulation with IFN-gamma plus lipopolysaccharide (LPS). Farnesol inhibited production of both IL-12 p40 and p70 from IFN-gamma/LPS-stimulated macrophages. Therefore, the role of farnesol in systemic candidiasis is likely due to its ability to inhibit the critical Th1 cytokines IFN-gamma and IL-12 and perhaps to enhance a Th2 cytokine, IL-5.
Collapse
|
44
|
Spellberg BJ, Collins M, Avanesian V, Gomez M, Edwards JE, Cogle C, Applebaum D, Fu Y, Ibrahim AS. Optimization of a myeloid cell transfusion strategy for infected neutropenic hosts. J Leukoc Biol 2006; 81:632-41. [PMID: 17158608 DOI: 10.1189/jlb.0906549] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Although granulocyte transfusion is a logical, therapeutic option for neutropenic patients with refractory infections, significant technical barriers have prevented its widespread use. A novel phagocyte transfusion strategy has been developed based on activation of a human myeloid cell line HL-60. To further define the potential for HL-60 cells to recapitulate white cell transfusions, a shortened duration of activation was evaluated, facile quality control markers were defined, and the impact of low-dose irradiation on cell function was determined. Three days of activation resulted in increased cell viability and in vitro candidacidal capacity but with slightly higher cell replication compared with 7 days of activation. Cell viability and several flow cytometric measurements were accurate, quality control markers for HL-60 activation. In combination with activation, low-dose irradiation abrogated replication while sparing the candidacidal effects of the HL-60 cells. Infusion of irradiated, activated HL-60 cells improved survival of neutropenic, candidemic mice significantly. In summary, activated, irradiated HL-60 cells are microbicidal, have virtually no replicative capacity, and are safe and effective at protecting neutropenic mice against an otherwise 100% fatal candidal infection. With continued development, this strategy to recapitulate neutrophil functions has the potential to serve as an effective alternative to granulocyte transfusions.
Collapse
Affiliation(s)
- Brad J Spellberg
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA 90502, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Cheng S, Clancy CJ, Zhang Z, Hao B, Wang W, Iczkowski KA, Pfaller MA, Nguyen MH. Uncoupling of oxidative phosphorylation enables Candida albicans to resist killing by phagocytes and persist in tissue. Cell Microbiol 2006; 9:492-501. [PMID: 16987332 DOI: 10.1111/j.1462-5822.2006.00805.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
After five serial passages of Candida albicans SC5314 through murine spleens by intravenous inoculation, we recovered a respiratory mutant (strain P5) that exhibited reduced colony size, stunted growth in glucose-deficient media, increased oxygen consumption and defective carbohydrate assimilation. Strain P5 was indistinguishable from SC5314 by DNA typing methods, but had a greater concentration of mitochondria by SYTO18 staining. Treatment with various inhibitors demonstrated that strain P5's electron transport chain was intact and oxidative phosphorylation was uncoupled. During disseminated candidiasis, the mutant did not kill mice or cause extensive damage to kidneys. The burden of strain P5 within kidneys on the first 3 days of disseminated candidiasis was significantly reduced. By days 28 and 60, it was similar to that at the time of death among mice infected with SC5314, suggesting that the mutant persisted and proliferated without killing mice. Strain P5 was resistant to phagocytosis by neutrophils and macrophages. It was also significantly more resistant to paraquat, suggesting that it is able to neutralize reactive oxygen species. Our findings indicate that regulation of respiration influences the interaction between C. albicans and the host. Uncoupling of oxidative phosphorylation might be a mechanism by which the organism adapts to stressful host environments.
Collapse
Affiliation(s)
- Shaoji Cheng
- The University of Florida College of Medicine, Gainesville, FL, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Filler SG. Candida-host cell receptor-ligand interactions. Curr Opin Microbiol 2006; 9:333-9. [PMID: 16837237 DOI: 10.1016/j.mib.2006.06.005] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2006] [Accepted: 06/19/2006] [Indexed: 11/27/2022]
Abstract
The interaction of Candida species with their cognate host receptors is a key factor in the pathogenesis of different types of candidiasis. The recognition of different forms of Candida albicans by Toll-like receptors 2 and 4 on mononuclear leukocytes has recently been discovered to determine the function and activity of regulatory T-cells, determine the balance of Type 1 and Type 2 cytokines and, thereby, influence the antifungal activity of both the innate and adaptive immune response. Different forms of C. albicans are also recognized by different lectins that are expressed on the surface macrophages. C. albicans and Candida glabrata express the ALS (agglutinin-like sequence) and EPA (epithelial adhesin) families of adhesins, respectively. A key difference between C. glabrata and C. albicans is that EPA expression in C. glabrata is governed by sub-telomeric silencing, whereas ALS expression in C. albicans is regulated by other mechanisms.
Collapse
Affiliation(s)
- Scott G Filler
- St Johns Cardiovascular Research Center, Division of Infectious Diseases, Department of Medicine, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA.
| |
Collapse
|
47
|
Spellberg BJ, Ibrahim AS, Avenissian V, Filler SG, Myers CL, Fu Y, Edwards JE. The anti-Candida albicans vaccine composed of the recombinant N terminus of Als1p reduces fungal burden and improves survival in both immunocompetent and immunocompromised mice. Infect Immun 2005; 73:6191-3. [PMID: 16113347 PMCID: PMC1231102 DOI: 10.1128/iai.73.9.6191-6193.2005] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We have previously shown that intraperitoneal vaccination with the recombinant N terminus of Als1p (rAls1p-N) modestly improves survival during murine disseminated candidiasis. We now report marked efficacy with subcutaneous rAls1p-N vaccination. Efficacy is retained in neutropenic and corticosteroid-treated mice. The rAls1p-N vaccine is a promising candidate for the prevention of invasive candidiasis.
Collapse
Affiliation(s)
- Brad J Spellberg
- Division of Infectious Diseases, Los Angeles Biomedical Institute at Harbor-UCLA Medical Center, 1124 W. Carson St., Torrance, CA 90502, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
Ito S, Pedras-Vasconcelos J, Klinman DM. CpG oligodeoxynucleotides increase the susceptibility of normal mice to infection by Candida albicans. Infect Immun 2005; 73:6154-6. [PMID: 16113339 PMCID: PMC1231092 DOI: 10.1128/iai.73.9.6154-6156.2005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Synthetic oligodeoxynucleotides containing CpG motifs trigger an innate immune response that typically increases host resistance to infection. Yet CpG treatment reduces the resistance of normal mice to Candida albicans infection. This effect is mediated by CpG-induced interleukin-12, indicating that CpG-dependent cytokine production may have adverse consequences for the host.
Collapse
Affiliation(s)
- Shuichi Ito
- Section of Retroviral Immunology, CBER/FDA, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
49
|
Spellberg BJ, Collins M, French SW, Edwards JE, Fu Y, Ibrahim AS. A phagocytic cell line markedly improves survival of infected neutropenic mice. J Leukoc Biol 2005; 78:338-44. [PMID: 15857941 DOI: 10.1189/jlb.0205072] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Disseminated candidiasis is a frequent infection in neutropenic patients, in whom it causes 50% mortality, despite antifungal therapy. As the duration of neutropenia is the strongest predictor of survival in neutropenic patients with invasive fungal infections, neutrophil transfusions are a logical, therapeutic option. However, significant technical barriers have prevented the clinical use of neutrophil transfusions. To overcome these barriers, we identified a human phagocytic cell line that could be administered to candidemic hosts in lieu of freshly harvested neutrophils. HL-60 cells killed Candida albicans in vitro. Activation of HL-60 cells with dimethyl sulfoxide and retinoic acid abrogated the cells' proliferation and augmented their killing of C. albicans. Administration of activated HL-60 cells to candidemic, neutropenic mice significantly improved survival (53% vs. 0%). Live HL-60 cells chemotaxed to sites of infection, phagocytized C. albicans, and reduced the fungal burden in key target organs. Although unactivated HL-60 cells also reduced tissue fungal burden in vivo, they did not improve survival as a result of their toxicity in infected mice. In contrast, no toxicity as a result of activated HL-60 cells was observed at up to 2 months of follow-up. To our knowledge, this is the first description of a cell line-based immunotherapy for an infectious disease. With further refinements, activated HL-60 cells have the potential to overcome the technical barriers to neutrophil transfusions.
Collapse
Affiliation(s)
- Brad J Spellberg
- Los Angeles Biomedical Institute, Harbor-UCLA Medical Center, Torrance, CA 90502, USA.
| | | | | | | | | | | |
Collapse
|
50
|
Ibrahim AS, Spellberg BJ, Avenissian V, Fu Y, Filler SG, Edwards JE. Vaccination with recombinant N-terminal domain of Als1p improves survival during murine disseminated candidiasis by enhancing cell-mediated, not humoral, immunity. Infect Immun 2005; 73:999-1005. [PMID: 15664943 PMCID: PMC547099 DOI: 10.1128/iai.73.2.999-1005.2005] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Candida spp. are opportunistic fungal pathogens that are among the most common causes of nosocomial bloodstream infections. The mortality attributable to disseminated candidiasis is 40 to 50% despite antifungal therapy. Clearly, new strategies are needed to prevent this life-threatening infection. Because risk factors for disseminated candidiasis are well defined and frequently of limited duration, vaccination is an appealing prophylactic strategy. We have identified a cell surface protein, Als1p, that mediates adherence of Candida albicans to a variety of human substrates and plastic. Here we report that immunizing BALB/c mice with the recombinant N-terminal domain of Als1p (rAls1p-N) improved survival during a subsequent challenge with a lethal inoculum of C. albicans. The protective 20-mug dose of rAls1p-N significantly increased Candida stimulation of Th1 splenocytes and increased in vivo delayed-type hypersensitivity. In contrast, antibody titers did not correlate with protection. Finally, the vaccine was not protective in T-cell-deficient mice but was protective in B-cell-deficient mice. These data indicate that the mechanism of action of the rAls1p-N vaccine is stimulation of cell-mediated, rather than humoral, immunity against C. albicans. The majority of efforts to date have focused on the development of passive immunization strategies to prevent or treat disseminated candidiasis. In contrast, our results provide proof of principle for vaccination with an adhesin of C. albicans and emphasize the potential for cell-mediated immune modulation as a prophylactic or therapeutic strategy against disseminated candidiasis.
Collapse
Affiliation(s)
- Ashraf S Ibrahim
- Division of Infectious Diseases, Los Angeles Biomedical Institute at Harbor-UCLA Medical Center, 1124 W. Carson Street, Torrance, CA 90502, USA.
| | | | | | | | | | | |
Collapse
|