1
|
Blair BA, Bragdon E, Dhillon G, Baker N, Stasiak L, Muthig M, Miramon P, Lorenz MC, Wheeler RT. Forward genetic screen in zebrafish identifies new fungal regulators that limit host-protective Candida-innate immune interaction. mBio 2025; 16:e0052925. [PMID: 40172223 PMCID: PMC12077120 DOI: 10.1128/mbio.00529-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 04/04/2025] Open
Abstract
Candida is one of the most frequent causes of bloodstream infections, and our first line of defense against these invasive infections is the innate immune system. The early immune response is critical in controlling Candida albicans infection, but C. albicans has several strategies to evade host immune attack. Phagocytosis of C. albicans blocks hyphal growth, limiting host damage and virulence, but how C. albicans limits early recruitment and phagocytosis in vertebrate infection is poorly understood. To study innate immune evasion by intravital imaging, we utilized the transparent larval zebrafish infection model to screen 131 C. albicans mutants for altered virulence and phagocyte response. Infections with each of the seven hypovirulent mutants led to altered phagocyte recruitment and/or phagocytosis, falling into four categories. Of particular interest among these is NMD5, a predicted β-importin and newly identified virulence factor. The nmd5∆/∆ mutant fails to limit phagocytosis, and its virulence defects are eliminated when phagocyte activity is compromised, suggesting that its role in virulence is limited to immune evasion. These quantitative intravital imaging experiments are the first to document altered Candida-phagocyte interactions for several additional mutants and clearly distinguish recruitment from phagocytic uptake, suggesting that Candida modulates both events. This initial large-scale screen of individual C. albicans mutants in a vertebrate, coupled with high-resolution imaging of Candida-phagocyte interactions, provides a more nuanced view of how diverse mutations can lead to more effective phagocytosis, a key immune process that blocks germination and drives anti-fungal immunity. IMPORTANCE Candida albicans is part of the human microbial community and is a dangerous opportunistic pathogen, able to prevent its elimination by the host immune system. Although Candida avoids immune attack through several strategies, we still understand little about how it regulates when immune phagocytes get recruited to the infection site and when they engulf fungal cells. We tested over 130 selected Candida mutants for their ability to cause lethal infection and found several hypovirulent mutants, which provoked altered innate immune responses, resulting in lower overall inflammation and greater host survival. Of particular interest is NMD5, which acts to limit fungal phagocytosis and is predicted to regulate the activity of stress-associated transcription factors. Our high-content screening was enabled by modeling Candida infection in transparent vertebrate zebrafish larva. Our findings help us understand how Candida survives immune attack during commensal and pathogenic growth, and may eventually inform new strategies for controlling disease.
Collapse
Affiliation(s)
- Bailey A. Blair
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, Maine, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, USA
| | - Emma Bragdon
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, Maine, USA
| | - Gursimran Dhillon
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, Maine, USA
| | - Nnamdi Baker
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, Maine, USA
| | - Lena Stasiak
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, Maine, USA
| | - Mya Muthig
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, Maine, USA
| | - Pedro Miramon
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Michael C. Lorenz
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Robert T. Wheeler
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, Maine, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, USA
| |
Collapse
|
2
|
Puumala E, Nandakumar M, Yiu B, Stogios PJ, Strickland BG, Zarnowski R, Wang X, Williams NS, Savchenko A, Andes DR, Robbins N, Whitesell L, Willson TM, Cowen LE. Structure-guided optimization of small molecules targeting Yck2 as a strategy to combat Candida albicans. Nat Commun 2025; 16:2156. [PMID: 40038303 PMCID: PMC11880385 DOI: 10.1038/s41467-025-57346-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/19/2025] [Indexed: 03/06/2025] Open
Abstract
Candida albicans is the most common cause of life-threatening fungal infection in the developed world but remains a therapeutic challenge. Protein kinases have been rewarding drug targets across diverse indications but remain untapped for antifungal development. Previously, screening kinase inhibitors against C. albicans revealed a 2,3-aryl-pyrazolopyridine, GW461484A (GW), which targets casein kinase 1 (CK1) family member Yck2. Here, we report optimization of GW via two complementary approaches, synthesis of bioisosteres possessing an imidazo[1,2-a]pyridine core, and R-group substitution of GW's pyrazolo[1,5-a]pyridine core. Characterization of compounds reveals two 6-cyano derivatives with improved pharmacological properties that retain whole-cell bioactivity and selectivity for fungal Yck2 compared to human CK1α. Efficacy studies in mice indicate both analogs possess single-agent activity against C. albicans resistant to first-line echinocandin antifungals and potentiate non-curative echinocandin treatment. Results validate Yck2 as an antifungal target and encourage further development of inhibitors acting by this previously unexploited mode of action.
Collapse
Affiliation(s)
- Emily Puumala
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Meganathan Nandakumar
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bonnie Yiu
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Peter J Stogios
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
| | - Benjamin G Strickland
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Robert Zarnowski
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Xiaoyu Wang
- Department of Biochemistry, University of Texas Southwestern Medical School, Dallas, TX, USA
| | - Noelle S Williams
- Department of Biochemistry, University of Texas Southwestern Medical School, Dallas, TX, USA
| | - Alexei Savchenko
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Center for Structural Biology of Infectious Diseases (CSBID), Chicago, Illinois, USA
| | - David R Andes
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Luke Whitesell
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Timothy M Willson
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
3
|
Blair BA, Bragdon E, Dhillon G, Baker N, Stasiak L, Muthig M, Miramon P, Lorenz MC, Wheeler RT. Forward genetic screen in zebrafish identifies new fungal regulators that limit host-protective Candida-innate immune interaction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.14.638315. [PMID: 39990375 PMCID: PMC11844468 DOI: 10.1101/2025.02.14.638315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Candida is one of the most frequent causes of bloodstream infections, and our first line of defense against these invasive infections is the innate immune system. The early immune response is critical in controlling C. albicans infection, but C. albicans has several strategies to evade host immune attack. Phagocytosis of C. albicans blocks hyphal growth, limiting host damage and virulence, but how C. albicans limits early recruitment and phagocytosis in vertebrate infection is poorly understood. To study innate immune evasion by intravital imaging, we utilized the transparent larval zebrafish infection model to screen 131 C. albicans mutants for altered virulence and phagocyte response. Infections with each of seven hypovirulent mutants led to altered phagocyte recruitment and/or phagocytosis, falling into four categories. Of particular interest among these is NMD5, a predicted β-importin and newly-identified virulence factor. The nmd5∆/∆ mutant fails to limit phagocytosis and its virulence defects are eliminated when phagocyte activity is compromised, suggesting that its role in virulence is limited to immune evasion. These quantitative intravital imaging experiments are the first to document altered Candida-phagocyte interactions for several additional mutants, and clearly distinguish recruitment from phagocytic uptake, suggesting that Candida modulates both events. This initial large-scale screen of individual C. albicans mutants in a vertebrate, coupled with high-resolution imaging of Candida-phagocyte interactions, provides a more nuanced view of how diverse mutations can lead to more effective phagocytosis, a key immune process which blocks germination and drives anti-fungal immunity.
Collapse
Affiliation(s)
- Bailey A. Blair
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME 04469
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469
| | - Emma Bragdon
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME 04469
| | - Gursimran Dhillon
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME 04469
| | - Nnamdi Baker
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME 04469
| | - Lena Stasiak
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME 04469
| | - Mya Muthig
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME 04469
| | - Pedro Miramon
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, USA
| | - Michael C. Lorenz
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, USA
| | - Robert T. Wheeler
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME 04469
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469
| |
Collapse
|
4
|
Cowen L, Puumala E, Nandakumar M, Yiu B, Stogios P, Strickland B, Zarnowski R, Wang X, Williams N, Savchenko A, Andes D, Robbins N, Whitesell L, Willson T. Structure-guided optimization of small molecules targeting the yeast casein kinase, Yck2, as a therapeutic strategy to combat Candida albicans. RESEARCH SQUARE 2025:rs.3.rs-5524306. [PMID: 39866870 PMCID: PMC11760248 DOI: 10.21203/rs.3.rs-5524306/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Candida albicans is the most common cause of life-threatening fungal infection in the developed world but remains a therapeutic challenge. Protein kinases have been rewarding drug targets across diverse indications but remain untapped for antifungal development. Previously, screening kinase inhibitors against C. albicans revealed a 2,3-aryl-pyrazolopyridine, GW461484A (GW), which targets casein kinase 1 (CK1) family member Yck2. Here, we report optimization of GW via two complementary approaches, synthesis of bioisosteres possessing an imidazo[1,2-a]pyridine core, and R-group substitution of GW's pyrazolo[1,5-a]pyridine core. Characterization of compounds synthesized revealed two 6-cyano derivatives with improved pharmacological properties that retained whole-cell bioactivity and selectivity for fungal Yck2 compared to human CK1α. Efficacy studies in mice indicated both analogs possess single-agent activity against C. albicans resistant to first-line echinocandin antifungals and potentiate non-curative echinocandin treatment. Results validate Yck2 as an antifungal target and encourage further development of inhibitors acting by this previously unexploited mode of action.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Noelle Williams
- The University of Texas Southwestern Medical Center at Dallas
| | | | | | | | | | | |
Collapse
|
5
|
Cheng M, Liu S, Zhu M, Li M, Yu Q. Adhesin Antibody-Grafted Mesoporous Silica Nanoparticles Suppress Immune Escape for Treatment of Fungal Systemic Infection. Molecules 2024; 29:4547. [PMID: 39407477 PMCID: PMC11478059 DOI: 10.3390/molecules29194547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/12/2024] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
Life-threatening systemic fungal infections caused by Candida albicans are significant contributors to clinical mortality, particularly among cancer patients and immunosuppressed individuals. The evasion of the immune response facilitated by fungal surface components enables fungal pathogens to evade macrophage attacks and to establish successful infections. This study developed a mesoporous silica nanoplatform, i.e., MSNP-EAP1Ab, which is composed of mesoporous silica nanoparticles grafted with the antibody of C. albicans surface adhesin Eap1. The activity of MSNP-EAP1Ab against C. albicans immune escape and infection was then evaluated by using the cell interaction model and the mouse systemic infection model. During interaction between C. albicans cells and macrophages, MSNP-EAP1Ab significantly inhibited fungal immune escape, leading to the enhanced phagocytosis of fungal cells by macrophages, with phagocytosis rates increasing from less than 8% to 14%. Furthermore, after treatment of the C. albicans-infected mice, MSNP-EAP1Ab drastically prolonged the mouse survival time and decreased the kidney fungal burden from >30,0000 CFU/g kidney to <100 CFU/g kidney, indicating the rapid recognition and killing of the pathogens by immune cells. Moreover, MSNP-EAP1Ab attenuated kidney tissue inflammation, with remarkable attenuation of renal immune cell accumulation. This study presents an innovative nanoplatform that targets the C. albicans adhesin, offering a promising approach for combatting systemic fungal infections.
Collapse
Affiliation(s)
- Mengjuan Cheng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, College of Life Sciences, Nankai University, Tianjin 300071, China; (M.C.)
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Suke Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, College of Life Sciences, Nankai University, Tianjin 300071, China; (M.C.)
| | - Mengsen Zhu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, College of Life Sciences, Nankai University, Tianjin 300071, China; (M.C.)
| | - Mingchun Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, College of Life Sciences, Nankai University, Tianjin 300071, China; (M.C.)
| | - Qilin Yu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, College of Life Sciences, Nankai University, Tianjin 300071, China; (M.C.)
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
6
|
Gao Y, Cao Q, Xiao Y, Wu Y, Ding L, Huang H, Li Y, Yang J, Meng L. The progress and future of the treatment of Candida albicans infections based on nanotechnology. J Nanobiotechnology 2024; 22:568. [PMID: 39285480 PMCID: PMC11406819 DOI: 10.1186/s12951-024-02841-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/07/2024] [Indexed: 09/19/2024] Open
Abstract
Systemic infection with Candida albicans poses a significant risk for people with weakened immune systems and carries a mortality rate of up to 60%. However, current therapeutic options have several limitations, including increasing drug tolerance, notable off-target effects, and severe adverse reactions. Over the past four decades, the progress in developing drugs to treat Candida albicans infections has been sluggish. This comprehensive review addresses the limitations of existing drugs and summarizes the efforts made toward redesigning and innovating existing or novel drugs through nanotechnology. The discussion explores the potential applications of nanomedicine in Candida albicans infections from four perspectives: nano-preparations for anti-biofilm therapy, innovative formulations of "old drugs" targeting the cell membrane and cell wall, reverse drug resistance therapy targeting subcellular organelles, and virulence deprivation therapy leveraging the unique polymorphism of Candida albicans. These therapeutic approaches are promising to address the above challenges and enhance the efficiency of drug development for Candida albicans infections. By harnessing nano-preparation technology to transform existing and preclinical drugs, novel therapeutic targets will be uncovered, providing effective solutions and broader horizons to improve patient survival rates.
Collapse
Affiliation(s)
- Yang Gao
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Qinyan Cao
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Yuyang Xiao
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Yue Wu
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Liang Ding
- Nanjing Stomatological Hospital, Nanjing, 210008, China
| | - He Huang
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Yanan Li
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China.
| | - Jingpeng Yang
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China.
| | - Lingtong Meng
- International Center for Synthetic Biology, School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China.
| |
Collapse
|
7
|
Ren J, Wu PP, Xue JH, Zhao WL, Zhu YH, Chen YY, Yang QJ, Luo Q, Cheng X, Bi EG. Discovery of an immunosuppressive functional metabolite from the insect-derived endophytic Aspergillus taichungensis SMU01. Fitoterapia 2024; 176:106007. [PMID: 38744384 DOI: 10.1016/j.fitote.2024.106007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/29/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
Three p-terphenyl metabolites (1-3), three indole-diterpenoids (4-6), an herbicide sesquiterpene (7), a flavonoid (8), and five other small molecules containing nitrogen (9-13) were isolated from the medicinal insect (Periplaneta americana)-derived endophytic Aspergillus taichungensis SMU01. Their chemical structures were elucidated on the basis of spectroscopic data and quantum chemical computational methods. Biological activity of these isolates in the differentiation of mouse CD4+ T cell subsets was evaluated. Importantly, metabolites 2 targeting JAK-STAT signaling pathway could hold potential benefits in maintaining peripheral immune homeostasis and alleviating the progression of autoimmune diseases.
Collapse
Affiliation(s)
- Jie Ren
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ping-Ping Wu
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Mediscine, Southern Medical University, Guangzhou 510515, China; School of Pharmaceutical Sciences, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Jia-Hao Xue
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Mediscine, Southern Medical University, Guangzhou 510515, China
| | - Wen-Li Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yi-Han Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yu-Yang Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Qian-Jun Yang
- Department of Stomatology, Jiangmen Central Hospital, Jiangmen 529000, China
| | - Qi Luo
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Mediscine, Southern Medical University, Guangzhou 510515, China.
| | - Xia Cheng
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Mediscine, Southern Medical University, Guangzhou 510515, China.
| | - En-Guang Bi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
8
|
Ashrafi S, Amini AA, Karimi P, Bagherian M, Adibzadeh Sereshgi MM, Asgarhalvaei F, Ahmadi K, Yazdi MH, Jahantigh HR, Mahdavi M, Sarrami Forooshani R. Candidiasis in breast cancer: Tumor progression or not? IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:1346-1356. [PMID: 39386227 PMCID: PMC11459349 DOI: 10.22038/ijbms.2024.75408.16379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 04/06/2024] [Indexed: 10/12/2024]
Abstract
Candida albicans is an "opportunistic fungal agent" in cancer patients that can become colonized in both mucosal and deep tissues and cause severe infections. Most evidence has shown that C. albicans can enhance the progress of different cancers by several mechanisms such as generating virulence factors, participation in endogenous production of pro-inflammatory mediators, and stimulating a wide range of immune cells in the host. The main idea of this review is to describe a range of Candida-used mechanisms that are important in candidiasis-associated malignant processes and cancer development, particularly breast cancer. This review intends to provide a detailed discussion on different regulatory mechanisms of C. albicans that undoubtedly help to open new therapeutic horizons of cancer therapy in patients with fungal infection. The current therapeutic approach is not fully effective in immunocompromised and cancer patients, and further studies are required to find new products with effective antifungal properties and minimal side effects to increase the susceptibility of opportunistic fungal infections to conventional antifungal agents. So, in this situation, a special therapy should be considered to control the infection and simultaneously have the most therapeutic index on tumor patients.
Collapse
Affiliation(s)
- Somayeh Ashrafi
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Recombinant Vaccine Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Microbiology, Faculty of Advanced Sciences & Technology, Tehran Medical Sciences, Islamic Azad University, (IAUPS), Tehran, Iran
- These authors contributed eqully to this work
| | - Abbas Ali Amini
- Department of Immunology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Cancer and Immunology Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Pegah Karimi
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Department of Biochemistry, Faculty of Basic Sciences, Islamic Azad University, Central Tehran Branch, Tehran, Iran
- These authors contributed eqully to this work
| | - Maryam Bagherian
- Department of Hematology and Oncology and Stem Cell Transplantation, Firoozgar Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Fatemeh Asgarhalvaei
- Department of Microbiology, Faculty of Advanced Sciences & Technology, Tehran Medical Sciences, Islamic Azad University, (IAUPS), Tehran, Iran
| | - Khadijeh Ahmadi
- Infectious and Tropical Diseases Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mohammad Hossein Yazdi
- Recombinant Vaccine Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Biotechnology Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Immunotherapy Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Jahantigh
- Animal Health and Zoonosis PhD Course, Department of Veterinary Medicine, University of Bari, Bari, Italy
| | - Mehdi Mahdavi
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Recombinant Vaccine Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Immunotherapy Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Ramin Sarrami Forooshani
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| |
Collapse
|
9
|
Jusuf S, Dong PT. Chromophore-Targeting Precision Antimicrobial Phototherapy. Cells 2023; 12:2664. [PMID: 37998399 PMCID: PMC10670386 DOI: 10.3390/cells12222664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/11/2023] [Accepted: 11/18/2023] [Indexed: 11/25/2023] Open
Abstract
Phototherapy, encompassing the utilization of both natural and artificial light, has emerged as a dependable and non-invasive strategy for addressing a diverse range of illnesses, diseases, and infections. This therapeutic approach, primarily known for its efficacy in treating skin infections, such as herpes and acne lesions, involves the synergistic use of specific light wavelengths and photosensitizers, like methylene blue. Photodynamic therapy, as it is termed, relies on the generation of antimicrobial reactive oxygen species (ROS) through the interaction between light and externally applied photosensitizers. Recent research, however, has highlighted the intrinsic antimicrobial properties of light itself, marking a paradigm shift in focus from exogenous agents to the inherent photosensitivity of molecules found naturally within pathogens. Chemical analyses have identified specific organic molecular structures and systems, including protoporphyrins and conjugated C=C bonds, as pivotal components in molecular photosensitivity. Given the prevalence of these systems in organic life forms, there is an urgent need to investigate the potential impact of phototherapy on individual molecules expressed within pathogens and discern their contributions to the antimicrobial effects of light. This review delves into the recently unveiled key molecular targets of phototherapy, offering insights into their potential downstream implications and therapeutic applications. By shedding light on these fundamental molecular mechanisms, we aim to advance our understanding of phototherapy's broader therapeutic potential and contribute to the development of innovative treatments for a wide array of microbial infections and diseases.
Collapse
Affiliation(s)
- Sebastian Jusuf
- Division of Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA;
| | - Pu-Ting Dong
- Department of Microbiology, The Forsyth Institute, Boston, MA 02142, USA
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA
| |
Collapse
|
10
|
Miao Y, Ding T, Liu Y, Zhou X, Du J. The Yeast and Hypha Phases of Candida krusei Induce the Apoptosis of Bovine Mammary Epithelial Cells via Distinct Signaling Pathways. Animals (Basel) 2023; 13:3222. [PMID: 37893947 PMCID: PMC10603689 DOI: 10.3390/ani13203222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Infection with Candida spp. is a significant cause of bovine mastitis globally. We previously found that C. krusei was the main pathogen causing mycotic mastitis in dairy cows in Yinchuan, Ningxia, China. However, whether the infection of this pathogen could induce apoptosis in BMECs remained unclear. In this report, we explored the apoptosis and underlying mechanism of BMECs induced by C. krusei yeast and hypha phases using a pathogen/host cell co-culture model. Our results revealed that both the yeast and hypha phases of C. krusei could induce BMEC apoptosis; however, the yeast phase induced more cell apoptosis than the hypha phase, as assessed via electronic microscopy and flow cytometry assays. This finding was further corroborated via the measurement of the mitochondrial membrane potential (MMP) and the TUNEL test. Infection by both the yeast and hypha phases of C. krusei greatly induced the expression of proteins associated with cell death pathways and important components of toll-like receptor (TLR) signaling, including TLR2 and TLR4 receptors, as determined via a Western blotting assay. BMECs mainly underwent apoptosis after infection by the C. krusei yeast phase through a mitochondrial pathway. Meanwhile, BMEC apoptosis induced by the C. krusei hypha phase was regulated by a death ligand/receptor pathway. In addition, C. krusei-induced BMEC apoptosis was regulated by both the TLR2/ERK and JNK/ERK signaling pathways. These data suggest that the yeast phase and hypha phase of C. krusei induce BMEC apoptosis through distinct cell signaling pathways. This study represents a unique perspective on the molecular processes underlying BMEC apoptosis in response to C. krusei infection.
Collapse
Affiliation(s)
- Yuhang Miao
- College of Life Science, Ningxia University, Yinchuan 750021, China; (Y.M.); (T.D.); (Y.L.)
- Key Laboratory of the Ministry of Education for the Conservation and Utilization of Special Biological Resources of Western China, Ningxia University, Yinchuan 750021, China
| | - Tao Ding
- College of Life Science, Ningxia University, Yinchuan 750021, China; (Y.M.); (T.D.); (Y.L.)
- Key Laboratory of the Ministry of Education for the Conservation and Utilization of Special Biological Resources of Western China, Ningxia University, Yinchuan 750021, China
| | - Yang Liu
- College of Life Science, Ningxia University, Yinchuan 750021, China; (Y.M.); (T.D.); (Y.L.)
- Key Laboratory of the Ministry of Education for the Conservation and Utilization of Special Biological Resources of Western China, Ningxia University, Yinchuan 750021, China
| | - Xuezhang Zhou
- College of Life Science, Ningxia University, Yinchuan 750021, China; (Y.M.); (T.D.); (Y.L.)
- Key Laboratory of the Ministry of Education for the Conservation and Utilization of Special Biological Resources of Western China, Ningxia University, Yinchuan 750021, China
| | - Jun Du
- College of Life Science, Ningxia University, Yinchuan 750021, China; (Y.M.); (T.D.); (Y.L.)
- Key Laboratory of the Ministry of Education for the Conservation and Utilization of Special Biological Resources of Western China, Ningxia University, Yinchuan 750021, China
| |
Collapse
|
11
|
Arafa SH, Elbanna K, Osman GEH, Abulreesh HH. Candida diagnostic techniques: a review. JOURNAL OF UMM AL-QURA UNIVERSITY FOR APPLIED SCIENCES 2023; 9:360-377. [DOI: 10.1007/s43994-023-00049-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/27/2023] [Indexed: 01/03/2025]
Abstract
AbstractFungal infections (mycoses) represent a major health issue in humans. They have emerged as a global concern for medical professionals by causing high morbidity and mortality. Fungal infections approximately impact one billion individuals per annum and account for 1.6 million deaths. The diagnosis of Candida infections is a challenging task. Laboratory-based Candida species identification techniques (molecular, commercial, and conventional) have been reviewed and summarized. This review aims to discuss the mycoses history, taxonomy, pathogenicity, and virulence characteristics.
Collapse
|
12
|
Lange T, Kasper L, Gresnigt MS, Brunke S, Hube B. "Under Pressure" - How fungi evade, exploit, and modulate cells of the innate immune system. Semin Immunol 2023; 66:101738. [PMID: 36878023 PMCID: PMC10109127 DOI: 10.1016/j.smim.2023.101738] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Indexed: 03/06/2023]
Abstract
The human immune system uses an arsenal of effector mechanisms to prevent and counteract infections. Yet, some fungal species are extremely successful as human pathogens, which can be attributed to a wide variety of strategies by which these fungi evade, exploit, and modulate the immune system. These fungal pathogens normally are either harmless commensals or environmental fungi. In this review we discuss how commensalism, but also life in an environmental niche without human contact, can drive the evolution of diverse and specialized immune evasion mechanisms. Correspondingly, we discuss the mechanisms contributing to the ability of these fungi to cause superficial to life-threatening infections.
Collapse
Affiliation(s)
- Theresa Lange
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany
| | - Lydia Kasper
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany
| | - Mark S Gresnigt
- Junior Research Group Adaptive Pathogenicity Strategies, Hans Knoell Institute, Jena, Germany
| | - Sascha Brunke
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany; Institute of Microbiology, Friedrich Schiller University, Jena, Germany.
| |
Collapse
|
13
|
Godoy P, Darlington PJ, Whiteway M. Genetic Screening of Candida albicans Inactivation Mutants Identifies New Genes Involved in Macrophage-Fungal Cell Interactions. Front Microbiol 2022; 13:833655. [PMID: 35450285 PMCID: PMC9016338 DOI: 10.3389/fmicb.2022.833655] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 03/08/2022] [Indexed: 11/13/2022] Open
Abstract
Candida albicans, an important fungal pathogen of humans, displays different morphologies, such as yeast, pseudo-hyphae and hyphae, which are recognized unequally by phagocytic cells of the innate immune response. Once C. albicans cells invade host tissues, immune cells such as macrophages are attracted to the site of infection and activated to recognize, engulf and kill the pathogen. We have investigated this fungal cell-macrophage interface by using high-throughput screening of the C. albicans GRACE library to identify genes that can influence this interaction and modify the kinetics of engulfment. Compared with the wild-type (WT) strain, we identified generally faster rates of engulfment for those fungal strains with constitutive pseudo-hyphal and hyphal phenotypes, whereas yeast-form-locked strains showed a reduced and delayed recognition and internalization by macrophages. We identified a number of GRACE strains that showed normal morphological development but exhibited different recognition and engulfment kinetics by cultured macrophages and characterized two mutants that modified interactions with the murine and human-derived macrophages. One mutant inactivated an uncharacterized C. albicans open reading frame that is the ortholog of S. cerevisiae OPY1, the other inactivated CaKRE1. The modified interaction was monitored during a 4 h co-culture. Early in the interaction, both opy1 and kre1 mutant strains showed reduced recognition and engulfment rates by macrophages when compared with WT cells. At fungal germ tube initiation, the engulfment kinetics increased for both mutants and WT cells, however the WT cells still showed a higher internalization by macrophages up to 2 h of interaction. Subsequently, between 2 and 4 h of the interaction, when most macrophages contain engulfed fungal cells, the engulfment kinetics increased for the opy1 mutant and further decreased for the kre1 mutant compared with Ca-WT. It appears that fungal morphology influences macrophage association with C. albicans cells and that both OPY1 and KRE1 play roles in the interaction of the fungal cells with phagocytes.
Collapse
Affiliation(s)
- Pablo Godoy
- Centre of Structural and Functional Genomics, Biology Department, Concordia University - Loyola Campus, Montreal, QC, Canada.,Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, Canada
| | - Peter John Darlington
- Perform Centre, Department of Health, Kinesiology and Applied Physiology, Montreal, QC, Canada
| | - Malcolm Whiteway
- Centre of Structural and Functional Genomics, Biology Department, Concordia University - Loyola Campus, Montreal, QC, Canada
| |
Collapse
|
14
|
Feng Y, Bian S, Pang Z, Wen Y, Calderone R, Li D, Shi D. Deletion of Non-histidine Domains of Histidine Kinase CHK1 Diminishes the Infectivity of Candida albicans in an Oral Mucosal Model. Front Microbiol 2022; 13:855651. [PMID: 35531278 PMCID: PMC9069115 DOI: 10.3389/fmicb.2022.855651] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/11/2022] [Indexed: 11/20/2022] Open
Abstract
Objectives The histidine kinase (HK) CHK1 and other protein kinases in Candida albicans are key players in the development of hyphae. This study is designed to determine the functional roles of the S_Tkc domain (protein kinase) and the GAF domain of C. albicans CHK1 in hyphal formation and mucosal invasion. Methods The domain mutants CHK25 (ΔS_TkcCHK1/Δchk1) and CHK26 (ΔS_TkcΔgafCHK1/Δchk1) were first constructed by the his1-URA3-his1 method and confirmed by sequencing and Southern blots. A mouse tongue infection model was used to evaluate the hyphal invasion and fungal loads in each domain mutant, full-gene deletion mutant CHK21 (chk1Δ/chk1Δ), re-constituted strain CHK23 (chk1Δ/CHK1), and wild type (WT) from day 1 to day 5. The degree of invasion and damage to the oral mucosa of mice in each strain-infected group was evaluated in vivo and compared with germ tube rate and hyphal formation in vitro. Result When compared with severe mucosal damage and massive hyphal formation in WT- or CHK23-infected mouse tongues, the deletion of S_Tkc domain (CHK25) caused mild mucosal damage, and fungal invasion was eliminated as we observed in full-gene mutant CHK21. However, the deletion of S_Tkc and GAF (CHK26) partially restored the hyphal invasion and mucosal tissue damage that were exhibited in WT and CHK23. Regardless of the in vivo results, the decreased hyphal formation and germ tube in vitro were less apparent and quite similar between CHK25 and CHK26, especially at the late stage of the log phase where CHK26 was closer to WT and CHK23. However, growth defect and hyphal impairment of both domain mutants were similar to CHK21 in the early stages. Conclusion Our data suggest that both protein kinase (S_Tkc) and GAF domains in C. albicans CHK1 are required for hyphal invasiveness in mucosal tissue. The appropriate initiation of cell growth and hyphal formation at the lag phase is likely mediated by these two functional domains of CHK1 to maintain in vivo infectivity of C. albicans.
Collapse
Affiliation(s)
- Yahui Feng
- College of Clinical Medicine, Jining Medical University, Jining, China
| | - Shaodong Bian
- Laboratory of Medical Mycology, Jining No. 1 People’s Hospital, Jining, China
| | - Zhiping Pang
- Laboratory of Medical Mycology, Jining No. 1 People’s Hospital, Jining, China
| | - Yiyang Wen
- Department of Pathology, Jining No. 1 People’s Hospital, Jining, China
| | - Richard Calderone
- Department of Microbiology/Immunology, Georgetown University Medical Center, Washington, DC, United States
| | - Dongmei Li
- Department of Microbiology/Immunology, Georgetown University Medical Center, Washington, DC, United States
- *Correspondence: Dongmei Li,
| | - Dongmei Shi
- Laboratory of Medical Mycology, Jining No. 1 People’s Hospital, Jining, China
- Department of Dermatology, Jining No.1 People’s Hospital, Jining, China
- Dongmei Shi,
| |
Collapse
|
15
|
Gamal A, Kadry A, Elshaer M, Ghannoum MA, Department of Dermatology, Case Western Reserve University, Cleveland, OH, USA, These authors have contributed equally to this work and share first authorship, Department of Dermatology, Case Western Reserve University, Cleveland, OH, USA, These authors have contributed equally to this work and share first authorship, Department of Clinical Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt, Department of Dermatology, Case Western Reserve University, Cleveland, OH, USA, Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA. Novel Antifungals for the Treatment of Vulvovaginal Candidiasis: Where Are We? Infect Dis (Lond) 2022. [DOI: 10.17925/id.2022.1.1.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Vulvovaginal candidiasis (VVC) is a common health-related issue and the second most common cause of vaginitis. Previously, azole antifungals were the mainstay of VVC treatment. Additionally, boric acid and nystatin have been used topically for management of VVC. Despite being effective and well tolerated by most patients, the use of azoles may be limited in some cases. Currently, two new antifungal agents have received US Food and Drug Administration approval for use in the management of VVC. In this article, we briefly review treatment regimens used for the management of VVC over the past decade, the newly approved agents and their possible clinical application, and future treatment considerations.
Collapse
|
16
|
Interactions of Both Pathogenic and Nonpathogenic CUG Clade Candida Species with Macrophages Share a Conserved Transcriptional Landscape. mBio 2021; 12:e0331721. [PMID: 34903044 PMCID: PMC8669484 DOI: 10.1128/mbio.03317-21] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Candida species are a leading cause of opportunistic, hospital-associated bloodstream infections with high mortality rates, typically in immunocompromised patients. Several species, including Candida albicans, the most prevalent cause of infection, belong to the monophyletic CUG clade of yeasts. Innate immune cells such as macrophages are crucial for controlling infection, and C. albicans responds to phagocytosis by a coordinated induction of pathways involved in catabolism of nonglucose carbon sources, termed alternative carbon metabolism, which together are essential for virulence. However, the interactions of other CUG clade species with macrophages have not been characterized. Here, we analyzed transcriptional responses to macrophage phagocytosis by six Candida species across a range of virulence and clinical importance. We define a core induced response common to pathogenic and nonpathogenic species alike, heavily weighted to alternative carbon metabolism. One prominent pathogen, Candida parapsilosis, showed species-specific expansion of phagocytosis-responsive genes, particularly metabolite transporters. C. albicans and Candida tropicalis, the other prominent pathogens, also had species-specific responses, but these were largely comprised of functionally uncharacterized genes. Transcriptional analysis of macrophages also demonstrated highly correlated proinflammatory transcriptional responses to different Candida species that were largely independent of fungal viability, suggesting that this response is driven by recognition of conserved cell wall components. This study significantly broadens our understanding of host interactions in CUG clade species, demonstrating that although metabolic plasticity is crucial for virulence in Candida, it alone is not sufficient to confer pathogenicity. Instead, we identify sets of mostly uncharacterized genes that may explain the evolution of pathogenicity.
Collapse
|
17
|
Lok B, Adam MAA, Kamal LZM, Chukwudi NA, Sandai R, Sandai D. The assimilation of different carbon sources in Candida albicans: Fitness and pathogenicity. Med Mycol 2021; 59:115-125. [PMID: 32944760 DOI: 10.1093/mmy/myaa080] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 08/07/2020] [Accepted: 08/14/2020] [Indexed: 01/31/2023] Open
Abstract
Candida albicans is a commensal yeast commonly found on the skin and in the body. However, in immunocompromised individuals, the fungi could cause local and systemic infections. The carbon source available plays an important role in the establishment of C. albicans infections. The fungi's ability to assimilate a variety of carbon sources plays a vital role in its colonization, and by extension, its fitness and pathogenicity, as it often inhabits niches that are glucose-limited but rich in alternative carbon sources. A difference in carbon sources affect the growth and mating of C. albicans, which contributes to its pathogenicity as proliferation helps the fungi colonize its environment. The carbon source also affects its metabolism and signaling pathways, which are integral parts of the fungi's fitness and pathogenicity. As a big percentage of the carbon assimilated by C. albicans goes to cell wall biogenesis, the availability of different carbon sources will result in cell walls with variations in rigidity, adhesion, and surface hydrophobicity. In addition to the biofilm formation of the fungi, the carbon source also influences whether the fungi grow in yeast- or mycelial-form. Both forms play different roles in C. albicans's infection process. A better understanding of the role of the carbon sources in C. albicans's pathogenicity would contribute to more effective treatment solutions for fungal infections.
Collapse
Affiliation(s)
- Bronwyn Lok
- Infectomics Cluster, Advance Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| | - Mowaffaq Adam Ahmad Adam
- Infectomics Cluster, Advance Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| | - Laina Zarisa Mohd Kamal
- Infectomics Cluster, Advance Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| | - Nwakpa Anthony Chukwudi
- Infectomics Cluster, Advance Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| | - Rosline Sandai
- Faculty of Languages and Communication, Universiti Pendidikan Sultan Idris, Perak Darul Ridzuan, Malaysia
| | - Doblin Sandai
- Infectomics Cluster, Advance Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| |
Collapse
|
18
|
Machata S, Sreekantapuram S, Hünniger K, Kurzai O, Dunker C, Schubert K, Krüger W, Schulze-Richter B, Speth C, Rambach G, Jacobsen ID. Significant Differences in Host-Pathogen Interactions Between Murine and Human Whole Blood. Front Immunol 2021; 11:565869. [PMID: 33519798 PMCID: PMC7843371 DOI: 10.3389/fimmu.2020.565869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 11/30/2020] [Indexed: 01/01/2023] Open
Abstract
Murine infection models are widely used to study systemic candidiasis caused by C. albicans. Whole-blood models can help to elucidate host-pathogens interactions and have been used for several Candida species in human blood. We adapted the human whole-blood model to murine blood. Unlike human blood, murine blood was unable to reduce fungal burden and more substantial filamentation of C. albicans was observed. This coincided with less fungal association with leukocytes, especially neutrophils. The lower neutrophil number in murine blood only partially explains insufficient infection and filamentation control, as spiking with murine neutrophils had only limited effects on fungal killing. Furthermore, increased fungal survival is not mediated by enhanced filamentation, as a filament-deficient mutant was likewise not eliminated. We also observed host-dependent differences for interaction of platelets with C. albicans, showing enhanced platelet aggregation, adhesion and activation in murine blood. For human blood, opsonization was shown to decrease platelet interaction suggesting that complement factors interfere with fungus-to-platelet binding. Our results reveal substantial differences between murine and human whole-blood models infected with C. albicans and thereby demonstrate limitations in the translatability of this ex vivo model between hosts.
Collapse
Affiliation(s)
- Silke Machata
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Sravya Sreekantapuram
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Kerstin Hünniger
- Research Group Fungal Septomics, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
- Institute for Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Oliver Kurzai
- Research Group Fungal Septomics, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
- Institute for Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Christine Dunker
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Katja Schubert
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Wibke Krüger
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Bianca Schulze-Richter
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Cornelia Speth
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Günter Rambach
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ilse D. Jacobsen
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
- Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
19
|
Roth C, Murray D, Scott A, Fu C, Averette AF, Sun S, Heitman J, Magwene PM. Pleiotropy and epistasis within and between signaling pathways defines the genetic architecture of fungal virulence. PLoS Genet 2021; 17:e1009313. [PMID: 33493169 PMCID: PMC7861560 DOI: 10.1371/journal.pgen.1009313] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 02/04/2021] [Accepted: 12/17/2020] [Indexed: 01/11/2023] Open
Abstract
Cryptococcal disease is estimated to affect nearly a quarter of a million people annually. Environmental isolates of Cryptococcus deneoformans, which make up 15 to 30% of clinical infections in temperate climates such as Europe, vary in their pathogenicity, ranging from benign to hyper-virulent. Key traits that contribute to virulence, such as the production of the pigment melanin, an extracellular polysaccharide capsule, and the ability to grow at human body temperature have been identified, yet little is known about the genetic basis of variation in such traits. Here we investigate the genetic basis of melanization, capsule size, thermal tolerance, oxidative stress resistance, and antifungal drug sensitivity using quantitative trait locus (QTL) mapping in progeny derived from a cross between two divergent C. deneoformans strains. Using a "function-valued" QTL analysis framework that exploits both time-series information and growth differences across multiple environments, we identified QTL for each of these virulence traits and drug susceptibility. For three QTL we identified the underlying genes and nucleotide differences that govern variation in virulence traits. One of these genes, RIC8, which encodes a regulator of cAMP-PKA signaling, contributes to variation in four virulence traits: melanization, capsule size, thermal tolerance, and resistance to oxidative stress. Two major effect QTL for amphotericin B resistance map to the genes SSK1 and SSK2, which encode key components of the HOG pathway, a fungal-specific signal transduction network that orchestrates cellular responses to osmotic and other stresses. We also discovered complex epistatic interactions within and between genes in the HOG and cAMP-PKA pathways that regulate antifungal drug resistance and resistance to oxidative stress. Our findings advance the understanding of virulence traits among diverse lineages of Cryptococcus, and highlight the role of genetic variation in key stress-responsive signaling pathways as a major contributor to phenotypic variation.
Collapse
Affiliation(s)
- Cullen Roth
- Department of Biology, Duke University, Durham, North Carolina, United States of America
- University Program in Genetics and Genomics, Duke University, Durham, North Carolina, United States of America
| | - Debra Murray
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - Alexandria Scott
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - Ci Fu
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Anna F. Averette
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Sheng Sun
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Joseph Heitman
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Paul M. Magwene
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| |
Collapse
|
20
|
Le PH, Nguyen DHK, Aburto-Medina A, Linklater DP, Crawford RJ, MacLaughlin S, Ivanova EP. Nanoscale Surface Roughness Influences Candida albicans Biofilm Formation. ACS APPLIED BIO MATERIALS 2020; 3:8581-8591. [PMID: 35019629 DOI: 10.1021/acsabm.0c00985] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The microbial contamination of surfaces presents a significant challenge due to the adverse effects associated with biofilm formation, particularly on implantable devices. Here, the attachment and biofilm formation of the opportunistic human pathogen, Candida albicans ATCC 10231, were studied on surfaces with decreasing magnitudes of nanoscale roughness. The nanoscale surface roughness of nonpolished titanium, polished titanium, and glass was characterized according to average surface roughness, skewness, and kurtosis. Nonpolished titanium, polished titanium, and glass possessed average surface roughness (Sa) values of 350, 20, and 2.5 nm; skewness (Sskw) values of 1.0, 4.0, and 1.0; and (Skur) values of 3.5, 16, and 4, respectively. These unique characteristics of the surface nanoarchitecture were found to play a key role in limiting C. albicans attachment and modulating the functional phenotypic changes associated with biofilm formation. Our results suggest that surfaces with a specific combination of surface topographical parameters could prevent the attachment and biofilm formation of C. albicans. After 7 days, the density of attached C. albicans cells was recorded to be 230, 70, and 220 cells mm-2 on nonpolished titanium, polished titanium, and glass surfaces, respectively. Despite achieving a very low attachment density, C. albicanscells were only observed to produce hyphae associated with biofilm formation on nonpolished titanium surfaces, possessing the highest degree of surface roughness (Sa = 350 nm). This study provides a more comprehensive picture of the impact of surface architectures on C. albicans attachment, which is beneficial for the design of antifungal surfaces.
Collapse
Affiliation(s)
- Phuc H Le
- School of Science, College of Science, Engineering and Health, RMIT University, Melbourne, Victoria 3000, Australia.,Australian Research Council Research Hub for Australian Steel Manufacturing, Wollongong, New South Wales 2500, Australia
| | - Duy H K Nguyen
- School of Science, College of Science, Engineering and Health, RMIT University, Melbourne, Victoria 3000, Australia
| | - Arturo Aburto-Medina
- School of Science, College of Science, Engineering and Health, RMIT University, Melbourne, Victoria 3000, Australia.,Australian Research Council Research Hub for Australian Steel Manufacturing, Wollongong, New South Wales 2500, Australia
| | - Denver P Linklater
- School of Science, College of Science, Engineering and Health, RMIT University, Melbourne, Victoria 3000, Australia
| | - Russell J Crawford
- School of Science, College of Science, Engineering and Health, RMIT University, Melbourne, Victoria 3000, Australia
| | | | - Elena P Ivanova
- School of Science, College of Science, Engineering and Health, RMIT University, Melbourne, Victoria 3000, Australia
| |
Collapse
|
21
|
Seegren PV, Downs TK, Stremska ME, Harper LR, Cao R, Olson RJ, Upchurch CM, Doyle CA, Kennedy J, Stipes EL, Leitinger N, Periasamy A, Desai BN. Mitochondrial Ca 2+ Signaling Is an Electrometabolic Switch to Fuel Phagosome Killing. Cell Rep 2020; 33:108411. [PMID: 33238121 PMCID: PMC7793167 DOI: 10.1016/j.celrep.2020.108411] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 07/30/2020] [Accepted: 10/28/2020] [Indexed: 11/17/2022] Open
Abstract
Phagocytes reallocate metabolic resources to kill engulfed pathogens, but the intracellular signals that rapidly switch the immunometabolic program necessary to fuel microbial killing are not understood. We report that macrophages use a fast two-step Ca2+ relay to meet the bioenergetic demands of phagosomal killing. Upon detection of a fungal pathogen, macrophages rapidly elevate cytosolic Ca2+ (phase 1), and by concurrently activating the mitochondrial Ca2+ (mCa2+) uniporter (MCU), they trigger a rapid influx of Ca2+ into the mitochondria (phase 2). mCa2+ signaling reprograms mitochondrial metabolism, at least in part, through the activation of pyruvate dehydrogenase (PDH). Deprived of mCa2+ signaling, Mcu−/− macrophages are deficient in phagosomal reactive oxygen species (ROS) production and defective at killing fungi. Mice lacking MCU in their myeloid cells are highly susceptible to disseminated candidiasis. In essence, this study reveals an elegant design principle that MCU-dependent Ca2+ signaling is an electrometabolic switch to fuel phagosome killing. The signaling mechanisms that rapidly reallocate metabolic resources to meet the bioenergetic demands of microbial killing are not understood. Seegren et al. show that mitochondrial Ca2+ signaling serves as a fast electrometabolic switch to fuel microbial killing by phagocytes. This study identifies the mitochondrial Ca2+ channel MCU as a critical component of cell-intrinsic antimicrobial responses.
Collapse
Affiliation(s)
- Philip V Seegren
- Pharmacology Department, University of Virginia, Pinn Hall, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA; Carter Immunology Center, University of Virginia, 345 Crispell r. MR-6, Charlottesville, VA 22908, USA
| | - Taylor K Downs
- Pharmacology Department, University of Virginia, Pinn Hall, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA; Carter Immunology Center, University of Virginia, 345 Crispell r. MR-6, Charlottesville, VA 22908, USA
| | - Marta E Stremska
- Carter Immunology Center, University of Virginia, 345 Crispell r. MR-6, Charlottesville, VA 22908, USA; Microbiology, Immunology, and Cancer Biology Department, University of Virginia, Pinn Hall, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA
| | - Logan R Harper
- Pharmacology Department, University of Virginia, Pinn Hall, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA
| | - Ruofan Cao
- The W.M. Keck Center for Cellular Imaging, University of Virginia, Physical and Life Sciences Building (PLSB), 90 Geldard Drive, Charlottesville, VA 22904, USA; Departments of Biology, University of Virginia, Physical and Life Sciences Building (PLSB), 90 Geldard Drive, Charlottesville, VA 22904, USA
| | - Rachel J Olson
- Pharmacology Department, University of Virginia, Pinn Hall, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA
| | - Clint M Upchurch
- Pharmacology Department, University of Virginia, Pinn Hall, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA; Carter Immunology Center, University of Virginia, 345 Crispell r. MR-6, Charlottesville, VA 22908, USA
| | - Catherine A Doyle
- Pharmacology Department, University of Virginia, Pinn Hall, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA; Carter Immunology Center, University of Virginia, 345 Crispell r. MR-6, Charlottesville, VA 22908, USA
| | - Joel Kennedy
- Pharmacology Department, University of Virginia, Pinn Hall, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA
| | - Eric L Stipes
- Pharmacology Department, University of Virginia, Pinn Hall, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA
| | - Norbert Leitinger
- Pharmacology Department, University of Virginia, Pinn Hall, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA; Carter Immunology Center, University of Virginia, 345 Crispell r. MR-6, Charlottesville, VA 22908, USA
| | - Ammasi Periasamy
- The W.M. Keck Center for Cellular Imaging, University of Virginia, Physical and Life Sciences Building (PLSB), 90 Geldard Drive, Charlottesville, VA 22904, USA; Departments of Biology, University of Virginia, Physical and Life Sciences Building (PLSB), 90 Geldard Drive, Charlottesville, VA 22904, USA
| | - Bimal N Desai
- Pharmacology Department, University of Virginia, Pinn Hall, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA; Carter Immunology Center, University of Virginia, 345 Crispell r. MR-6, Charlottesville, VA 22908, USA.
| |
Collapse
|
22
|
Laurian R, Jacot-des-Combes C, Bastian F, Dementhon K, Cotton P. Carbon metabolism snapshot by ddPCR during the early step of Candida albicans phagocytosis by macrophages. Pathog Dis 2020; 78:5780227. [PMID: 32129841 DOI: 10.1093/femspd/ftaa014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 03/03/2020] [Indexed: 01/08/2023] Open
Abstract
During Candida macrophage interactions, phagocytosed yeast cells feed in order to grow, develop hyphae and escape. Through numerous proteomic and transcriptomic studies, two metabolic phases have been described. A shift to a starvation mode is generally identified as early as one-hour post phagocytosis, followed by a glycolytic growth mode after C. albicans escaped from the macrophage. Healthy macrophages contain low amounts of glucose. To determine if this carbon source was sensed and metabolized by the pathogen, we explored the transcription level of a delimited set of key genes expressed in C. albicans cells during phagocytosis by macrophages, at an early stage of the interaction. This analysis was performed using a technical digital droplet PCR approach to quantify reliably the expression of carbon metabolic genes after 30 min of phagocytosis. Our data confirm the technique of digital droplet PCR for the detection of C. albicans transcripts using cells recovered after a short period of phagocytosis. At this stage, carbon metabolism is clearly oriented towards the use of alternative sources. However, the activation of high-affinity glucose transport system suggests that the low amount of glucose initially present in the macrophages is detected by the pathogen.
Collapse
Affiliation(s)
- Romain Laurian
- Génétique Moléculaire des Levures, UMR-CNRS 5240 Microbiologie Adaptation et Pathogénie, Université de Lyon-Université Lyon1, Lyon, France
| | - Cécile Jacot-des-Combes
- DTAMB, FR 3728 Bio-Environnement et Santé, Université de Lyon-Université Lyon1, Lyon, France
| | - Fabiola Bastian
- DTAMB, FR 3728 Bio-Environnement et Santé, Université de Lyon-Université Lyon1, Lyon, France
| | - Karine Dementhon
- Laboratoire de Microbiologie Fondamentale et Pathogénicité, UMR-CNRS 5234, Université de Bordeaux, Bordeaux, France
| | - Pascale Cotton
- Génétique Moléculaire des Levures, UMR-CNRS 5240 Microbiologie Adaptation et Pathogénie, Université de Lyon-Université Lyon1, Lyon, France
| |
Collapse
|
23
|
Khan MA, Khan A, Khan SH, Azam M, Khan MMU, Khalilullah H, Younus H. Coadministration of liposomal methylglyoxal increases the activity of amphotericin B against Candida albicans in leukopoenic mice. J Drug Target 2020; 29:78-87. [PMID: 32723117 DOI: 10.1080/1061186x.2020.1803333] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In this study, we investigated the therapeutic efficacy of a combination of liposomal amphotericin B (Lip-Amp B) and Methylglyoxal (Lip-MG) against Candida albicans in the leukopoenic mice. The antifungal efficacy of Lip-Amp B or Lip-MG or a combination of Lip-Amp B and Lip-MG was evaluated by the analysis of the survival rate and the fungal load in the treated mice. The immune-stimulatory effect of Lip-MG on macrophages was evaluated by analysing the secretion of proinflammatory cytokines. C. albicans infected mice treated at the doses of 1 and 2 mg/kg of Lip-Amp B showed 20% and 50% survival rates, respectively. Whereas the mice treated with free Amp B at the same doses died within 40 days of treatment. Interestingly, C. albicans infected mice treated with a combination of Lip-Amp B and Lip-MG had 70% survival rate on day 40 postinfection. Moreover, treatment of macrophages with Lip-MG increased their fungicidal activity and the secretion of proinflammatory cytokines, including TNF-α and IL-1β. These findings suggested that co-treatment with Lip-Amp B and Lip-MG had a synergistic effect and could be effective against C. albicans in immunocompromised subjects.
Collapse
Affiliation(s)
- Masood Alam Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Arif Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Shaheer Hasan Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Mohd Azam
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Mohd Masih Uzzaman Khan
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Qassim University, Saudi Arabia
| | - Habibullah Khalilullah
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Qassim University, Saudi Arabia
| | - Hina Younus
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
24
|
Zamudio KR, McDonald CA, Belasen AM. High Variability in Infection Mechanisms and Host Responses: A Review of Functional Genomic Studies of Amphibian Chytridiomycosis. HERPETOLOGICA 2020. [DOI: 10.1655/0018-0831-76.2.189] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Kelly R. Zamudio
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, NY 14853-2701, USA
| | - Cait A. McDonald
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, NY 14853-2701, USA
| | - Anat M. Belasen
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, NY 14853-2701, USA
| |
Collapse
|
25
|
Khan SH, Younus H, Allemailem KS, Almatroudi A, Alrumaihi F, Alruwetei AM, Alsahli MA, Khan A, Khan MA. Potential of Methylglyoxal-Conjugated Chitosan Nanoparticles in Treatment of Fluconazole-Resistant Candida albicans Infection in a Murine Model. Int J Nanomedicine 2020; 15:3681-3693. [PMID: 32547022 PMCID: PMC7261666 DOI: 10.2147/ijn.s249625] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/04/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Fungal infections are becoming more prevalent and threatening because of the continuous emergence of azole-resistant fungal infections. The present study was aimed to assess the activity of free Methylglyoxal (MG) or MG-conjugated chitosan nanoparticles (MGCN) against fluconazole-resistant Candida albicans. MATERIALS AND METHODS A novel formulation of MGCN was prepared and characterized to determine their size, shape and polydispersity index. Moreover, the efficacy of fluconazole or MG or MGCN was determined against intracellular C. albicans in macrophages and the systematic candidiasis in a murine model. The safety of MG or MGCN was tested in mice by analyzing the levels of hepatic and renal toxicity parameters. RESULTS Candida albicans did not respond to fluconazole, even at the highest dose of 20 mg/kg, whereas MG and MGCN effectively eliminated C. albicans from the macrophages and infected mice. Mice in the group treated with MGCN at a dose of 10 mg/kg exhibited a 90% survival rate and showed the lowest fungal load in the kidney, whereas the mice treated with free MG at the same dose exhibited 50% survival rate. Moreover, the administration of MG or MGCN did not induce any liver and kidney toxicity in the treated mice. CONCLUSION The findings of the present work suggest that MGCN may be proved a promising therapeutic formulation to treat azole-resistant C. albicans infections.
Collapse
Affiliation(s)
- Shaheer Hasan Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh202002, India
| | - Hina Younus
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh202002, India
| | - Khaled S Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah51452, Saudi Arabia
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah51452, Saudi Arabia
| | - Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah51452, Saudi Arabia
| | - Abdulmohsen M Alruwetei
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah51452, Saudi Arabia
| | - Mohammed A Alsahli
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah51452, Saudi Arabia
| | - Arif Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraidah, 51452, Saudi Arabia
| | - Masood Alam Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraidah, 51452, Saudi Arabia
| |
Collapse
|
26
|
Tarang S, Kesherwani V, LaTendresse B, Lindgren L, Rocha-Sanchez SM, Weston MD. In silico Design of a Multivalent Vaccine Against Candida albicans. Sci Rep 2020; 10:1066. [PMID: 31974431 PMCID: PMC6978452 DOI: 10.1038/s41598-020-57906-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 01/07/2020] [Indexed: 12/14/2022] Open
Abstract
Invasive candidiasis (IC) is the most common nosocomial infection and a leading cause of mycoses-related deaths. High-systemic toxicity and emergence of antifungal-resistant species warrant the development of newer preventive approaches against IC. Here, we have adopted an immunotherapeutic peptide vaccine-based approach, to enhance the body's immune response against invasive candida infections. Using computational tools, we screened the entire candida proteome (6030 proteins) and identified the most immunodominant HLA class I, HLA class II and B- cell epitopes. By further immunoinformatic analyses for enhanced vaccine efficacy, we selected the 18- most promising epitopes, which were joined together using molecular linkers to create a multivalent recombinant protein against Candida albicans (mvPC). To increase mvPC's immunogenicity, we added a synthetic adjuvant (RS09) to the mvPC design. The selected mvPC epitopes are homologous against all currently available annotated reference sequences of 22 C. albicans strains, thus offering a higher coverage and greater protective response. A major advantage of the current vaccine approach is mvPC's multivalent nature (recognizing multiple-epitopes), which is likely to provide enhanced protection against complex candida antigens. Here, we describe the computational analyses leading to mvPC design.
Collapse
Affiliation(s)
- Shikha Tarang
- Creighton University School of Dentistry, Department of Oral Biology, Omaha, NE, 68178, USA.
| | - Varun Kesherwani
- Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Blake LaTendresse
- Creighton University School of Dentistry, Department of Oral Biology, Omaha, NE, 68178, USA
| | - Laramie Lindgren
- Creighton University School of Dentistry, Department of Oral Biology, Omaha, NE, 68178, USA
| | - Sonia M Rocha-Sanchez
- Creighton University School of Dentistry, Department of Oral Biology, Omaha, NE, 68178, USA
| | - Michael D Weston
- Creighton University School of Dentistry, Department of Oral Biology, Omaha, NE, 68178, USA
| |
Collapse
|
27
|
Abstract
Candida albicans is a commensal as well as a pathogen of humans. C. albicans is able to mount a cellular response to a diverse range of external stimuli in the host and switch reversibly between the yeast and hyphal growth forms. Hyphal development is a key virulence determinant. Here, we studied how C. albicans senses different environmental signals to control its growth forms. Our study results suggest that robust hyphal development requires downregulation of two transcriptional repressors, Nrg1 and Sfl1. Acidic pH or cationic stress inhibits hyphal formation via stress-responsive kinases and Sfl1. Candida albicans is an important human pathogen responsible for causing both superficial and systemic infections. Its ability to switch from the yeast form to the hyphal growth form is required for its pathogenicity. Acidic pH inhibits hyphal initiation, but the nature of the mechanism for this inhibition is not completely clear. We show that acidic pH represses hyphal initiation independently of the temperature- and farnesol-mediated Nrg1 downregulation. Using a collection of transcription factor deletion mutants, we observed that the sfl1 mutant induced hyphae in acidic pH but not in farnesol at 37°C. Furthermore, transcription of hyphal regulators BRG1 and UME6 was not induced in wild-type (WT) cells but was induced in the sfl1 mutant during hyphal induction in acidic pH. Using the same screening conditions with the collection of kinase mutants, we found that deletions of the core stress response mitogen-activated protein (MAP) kinase HOG1 and its kinase PBS2, the cell wall stress MAP kinase MKC1, and the calcium/calmodulin-dependent kinase CMK1 allowed hyphal initiation in acidic pH. Furthermore, Hog1 phosphorylation induced by high osmotic stress also retarded hyphal initiation, and the effect was abolished in the sfl1 and three kinase mutants but was enhanced in the phosphatase mutant ptp2 ptp3. We also found functional associations among Cmk1, Hog1, and Sfl1 for cation stress. Our study results suggest that robust hyphal initiation requires downregulation of both Nrg1 and Sfl1 transcriptional repressors as well as timely BRG1 expression. Acidic pH and cationic stress retard hyphal initiation via the stress-responsive kinases and Sfl1. IMPORTANCECandida albicans is a commensal as well as a pathogen of humans. C. albicans is able to mount a cellular response to a diverse range of external stimuli in the host and switch reversibly between the yeast and hyphal growth forms. Hyphal development is a key virulence determinant. Here, we studied how C. albicans senses different environmental signals to control its growth forms. Our study results suggest that robust hyphal development requires downregulation of two transcriptional repressors, Nrg1 and Sfl1. Acidic pH or cationic stress inhibits hyphal formation via stress-responsive kinases and Sfl1.
Collapse
|
28
|
Wadhwa R, Pandey P, Gupta G, Aggarwal T, Kumar N, Mehta M, Satija S, Gulati M, Madan JR, Dureja H, Balusamy SR, Perumalsamy H, Maurya PK, Collet T, Tambuwala MM, Hansbro PM, Chellappan DK, Dua K. Emerging Complexity and the Need for Advanced Drug Delivery in Targeting Candida Species. Curr Top Med Chem 2019; 19:2593-2609. [DOI: 10.2174/1568026619666191026105308] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 07/15/2019] [Accepted: 09/07/2019] [Indexed: 02/07/2023]
Abstract
Background:Candida species are the important etiologic agents for candidiasis, the most prevalent cause of opportunistic fungal infections. Candida invasion results in mucosal to systemic infections through immune dysfunction and helps in further invasion and proliferation at several sites in the host. The host defence system utilizes a wide array of the cells, proteins and chemical signals that are distributed in blood and tissues which further constitute the innate and adaptive immune system. The lack of antifungal agents and their limited therapeutic effects have led to high mortality and morbidity related to such infections.Methods:The necessary information collated on this review has been gathered from various literature published from 1995 to 2019.Results:This article sheds light on novel drug delivery approaches to target the immunological axis for several Candida species (C. albicans, C. glabrata, C. parapsilosis, C. tropicalis, C. krusei, C. rugose, C. hemulonii, etc.).Conclusion:It is clear that the novel drug delivery approaches include vaccines, adoptive transfer of primed immune cells, recombinant cytokines, therapeutic antibodies, and nanoparticles, which have immunomodulatory effects. Such advancements in targeting various underpinning mechanisms using the concept of novel drug delivery will provide a new dimension to the fungal infection clinic particularly due to Candida species with improved patient compliance and lesser side effects. This advancement in knowledge can also be extended to target various other similar microbial species and infections.
Collapse
Affiliation(s)
- Ridhima Wadhwa
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Parijat Pandey
- Shri Baba Mastnath Institute of Pharmaceutical Sciences and Research, Baba Mastnath University, Rohtak 124001, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura 302 017, Jaipur, India
| | - Taru Aggarwal
- Amity Institute of Biotechnology, Amity University, Noida 201303, India
| | - Nitesh Kumar
- Amity Institute for Advanced Research & Studies (M&D), Amity University, Noida 201303, India
| | - Meenu Mehta
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar, Delhi G.T. Road (NH-1), Phagwara-144411, Punjab, India
| | - Saurabh Satija
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar, Delhi G.T. Road (NH-1), Phagwara-144411, Punjab, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar, Delhi G.T. Road (NH-1), Phagwara-144411, Punjab, India
| | - Jyotsna R. Madan
- Department of Pharmaceutics, Smt. Kashibai Navale College of Pharmacy, Kondhwa, Pune, 411048, Maharashtra, India
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharishi Dayanand University, Rohtak, Haryana 124001, India
| | - Sri R. Balusamy
- Department of Food Science and Biotechnology, Sejong University, Gwangjin-gu, Seoul, 05006, Korea
| | - Haribalan Perumalsamy
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin, 446-701, Korea
| | - Pawan K. Maurya
- Department of Biochemistry, Central University of Haryana, Jant-Pali, Mahendergarh District 123031, Haryana, India
| | - Trudi Collet
- Innovative Medicines Group, Institute of Health & Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Brisbane, Queensland 4059, Australia
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, County Londonderry, BT52 1SA, Northern Ireland, United Kingdom
| | - Philip M. Hansbro
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Kamal Dua
- School of Pharmaceutical Sciences, Shoolini University, Bajhol, Sultanpur, Solan, Himachal Pradesh 173 229, Australia
| |
Collapse
|
29
|
Naranjo‐Ortiz MA, Gabaldón T. Fungal evolution: major ecological adaptations and evolutionary transitions. Biol Rev Camb Philos Soc 2019; 94:1443-1476. [PMID: 31021528 PMCID: PMC6850671 DOI: 10.1111/brv.12510] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 03/10/2019] [Accepted: 03/13/2019] [Indexed: 12/13/2022]
Abstract
Fungi are a highly diverse group of heterotrophic eukaryotes characterized by the absence of phagotrophy and the presence of a chitinous cell wall. While unicellular fungi are far from rare, part of the evolutionary success of the group resides in their ability to grow indefinitely as a cylindrical multinucleated cell (hypha). Armed with these morphological traits and with an extremely high metabolical diversity, fungi have conquered numerous ecological niches and have shaped a whole world of interactions with other living organisms. Herein we survey the main evolutionary and ecological processes that have guided fungal diversity. We will first review the ecology and evolution of the zoosporic lineages and the process of terrestrialization, as one of the major evolutionary transitions in this kingdom. Several plausible scenarios have been proposed for fungal terrestralization and we here propose a new scenario, which considers icy environments as a transitory niche between water and emerged land. We then focus on exploring the main ecological relationships of Fungi with other organisms (other fungi, protozoans, animals and plants), as well as the origin of adaptations to certain specialized ecological niches within the group (lichens, black fungi and yeasts). Throughout this review we use an evolutionary and comparative-genomics perspective to understand fungal ecological diversity. Finally, we highlight the importance of genome-enabled inferences to envision plausible narratives and scenarios for important transitions.
Collapse
Affiliation(s)
- Miguel A. Naranjo‐Ortiz
- Department of Genomics and Bioinformatics, Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyDr. Aiguader 88, Barcelona08003Spain
| | - Toni Gabaldón
- Department of Genomics and Bioinformatics, Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyDr. Aiguader 88, Barcelona08003Spain
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF)08003BarcelonaSpain
- ICREA, Pg. Lluís Companys 2308010BarcelonaSpain
| |
Collapse
|
30
|
Arce Miranda JE, Baronetti JL, Sotomayor CE, Paraje MG. Oxidative and nitrosative stress responses during macrophage-Candida albicans biofilm interaction. Med Mycol 2019; 57:101-113. [PMID: 29294039 DOI: 10.1093/mmy/myx143] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 11/23/2017] [Indexed: 12/11/2022] Open
Abstract
Candida albicans is an important source of device-associated infection because of its capacity for biofilm formation. This yeast has the ability to form biofilms which favors the persistence of the infection. Furthermore, the innate immune response has a critical role in the control of these infections and macrophages (Mø) are vital to this process. An important fungicidal mechanism employed by Mø involves the generation of toxic reactive oxygen species (ROS) and reactive nitrogen intermediates (RNI). The interaction between biofilms and these immune cells, and the contribution of oxidative and nitrosative stress, that is determinant to the course of the infection, remains elusive. The aim of this study was to investigate this interaction. To this purpose, two models of Mø-biofilms contact, early (model 1) and mature (model 2) biofilms, were used; and the production of ROS, RNI and the oxidative stress response (OSR) were evaluated. We found that the presence of Mø decreased the biofilm formation at an early stage and increased the production of ROS and RNI, with activation of ORS (enzymatic and nonenzymatic). On the other hand, the interaction between mature biofilms and Mø resulted in an increasing biofilm formation, with low levels of RNI and ROS production and decrease of OSR. Dynamic interactions between Mø and fungal biofilms were also clearly evident from images obtained by confocal scanning laser microscopy. The prooxidant-antioxidant balance was different depending of C. albicans biofilms stages and likely acts as a signal over their formation in presence of Mø. These results may contribute to a better understanding of the immune-pathogenesis of C. albicans biofilm infections.
Collapse
Affiliation(s)
- Julio E Arce Miranda
- IMBIV-National Scientific and Technical Research Council (CONICET) and Department of Physiology, Faculty of Exact, Physical and Natural Sciences, National University of Córdoba, Córdoba, Argentina
| | - José L Baronetti
- IMBIV-National Scientific and Technical Research Council (CONICET) and Department of Physiology, Faculty of Exact, Physical and Natural Sciences, National University of Córdoba, Córdoba, Argentina
| | - Claudia E Sotomayor
- CIBICI-National Scientific and Technical Research Council (CONICET) and Department of Clinical Biochemistry, Faculty of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
| | - M Gabriela Paraje
- IMBIV-National Scientific and Technical Research Council (CONICET) and Department of Physiology, Faculty of Exact, Physical and Natural Sciences, National University of Córdoba, Córdoba, Argentina
| |
Collapse
|
31
|
Liao J, Pan B, Liao G, Zhao Q, Gao Y, Chai X, Zhuo X, Wu Q, Jiao B, Pan W, Guo Z. Synthesis and immunological studies of β-1,2-mannan-peptide conjugates as antifungal vaccines. Eur J Med Chem 2019; 173:250-260. [DOI: 10.1016/j.ejmech.2019.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/20/2019] [Accepted: 04/01/2019] [Indexed: 01/06/2023]
|
32
|
Lee SY, Chen HF, Yeh YC, Xue YP, Lan CY. The Transcription Factor Sfp1 Regulates the Oxidative Stress Response in Candida albicans. Microorganisms 2019; 7:E131. [PMID: 31091716 PMCID: PMC6560436 DOI: 10.3390/microorganisms7050131] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/07/2019] [Accepted: 05/09/2019] [Indexed: 12/15/2022] Open
Abstract
Candida albicans is a commensal that inhabits the skin and mucous membranes of humans. Because of the increasing immunocompromised population and the limited classes of antifungal drugs available, C. albicans has emerged as an important opportunistic pathogen with high mortality rates. During infection and therapy, C. albicans frequently encounters immune cells and antifungal drugs, many of which exert their antimicrobial activity by inducing the production of reactive oxygen species (ROS). Therefore, antioxidative capacity is important for the survival and pathogenesis of C. albicans. In this study, we characterized the roles of the zinc finger transcription factor Sfp1 in the oxidative stress response against C. albicans. A sfp1-deleted mutant was more resistant to oxidants and macrophage killing than wild-type C. albicans and processed an active oxidative stress response with the phosphorylation of the mitogen-activated protein kinase (MAPK) Hog1 and high CAP1 expression. Moreover, the sfp1-deleted mutant exhibited high expression levels of antioxidant genes in response to oxidative stress, resulting in a higher total antioxidant capacity, glutathione content, and glutathione peroxidase and superoxide dismutase enzyme activity than the wild-type C. albicans. Finally, the sfp1-deleted mutant was resistant to macrophage killing and ROS-generating antifungal drugs. Together, our findings provide a new understanding of the complex regulatory machinery in the C. albicans oxidative stress response.
Collapse
Affiliation(s)
- Shao-Yu Lee
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Hsueh-Fen Chen
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Ying-Chieh Yeh
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Yao-Peng Xue
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Chung-Yu Lan
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 30013, Taiwan.
- Department of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan.
| |
Collapse
|
33
|
Abstract
C. albicans is the most common cause of nosocomial fungal infection, and over 3 million people acquire life-threatening invasive fungal infections every year. Even if antifungal drugs exist, almost half of these patients will die. Despite this, fungi remain underestimated as pathogens. Our study uses quantitative biophysical approaches to demonstrate that yeast-to-hypha transition occurs within the nutrient-deprived, acidic phagosome and that alkalinization is a consequence, as opposed to the cause, of hyphal growth. Macrophages rely on phagosomal acidity to destroy engulfed microorganisms. To survive this hostile response, opportunistic fungi such as Candida albicans developed strategies to evade the acidic environment. C. albicans is polymorphic and able to convert from yeast to hyphae, and this transition is required to subvert the microbicidal activity of the phagosome. However, the phagosomal lumen, which is acidic and nutrient deprived, is believed to inhibit the yeast-to-hypha transition. To account for this apparent paradox, it was recently proposed that C. albicans produces ammonia that alkalinizes the phagosome, thus facilitating yeast-to-hypha transition. We reexamined the mechanism underlying phagosomal alkalinization by applying dual-wavelength ratiometric pH measurements. The phagosomal membrane was found to be highly permeable to ammonia, which is therefore unlikely to account for the pH elevation. Instead, we find that yeast-to-hypha transition begins within acidic phagosomes and that alkalinization is a consequence of proton leakage induced by excessive membrane distension caused by the expanding hypha.
Collapse
|
34
|
Abstract
Various fungi and bacteria can colonize in the brain and produce physical alterations seen in Alzheimer’s disease (AD). Environmental and genetic factors affect the occurrence of fungal colonization, and how fungi can grow, enter the brain, and interact with the innate immune system. The essence of AD development is the defeat of the innate immune system, whether through vulnerable patient health status or treatment that suppresses inflammation by suppressing the innate immune system. External and mechanical factors that lead to inflammation are a door for pathogenic opportunity. Current research associates the presence of fungi in the etiology of AD and is shown in cerebral tissue at autopsy. From the time of the discovery of AD, much speculation exists for an infective cause. Identifying any AD disease organism is obscured by processes that can take place over years. Amyloid protein deposits are generally considered to be evidence of an intrinsic response to stress or imbalance, but instead amyloid may be evidence of the innate immune response which exists to destroy fungal colonization through structural interference and cytotoxicity. Fungi can remain ensconced for a long time in niches or inside cells, and it is the harboring of fungi that leads to repeated reinfection and slow wider colonization that eventually leads to a grave outcome. Although many fungi and bacteria are associated with AD affected tissues, discussion here focuses on Candida albicans as the archetype of human fungal pathology because of its wide proliferation as a commensal fungus, extensive published research, numerous fungal morphologies, and majority proliferation in AD tissues.
Collapse
Affiliation(s)
- Bodo Parady
- Children's Hospital Oakland Research Institute, Oakland, CA, USA.,Visiting Scholar, University of California, Berkeley, Berkeley CA, USA
| |
Collapse
|
35
|
Zhang B, Yu Q, Huo D, Li J, Liang C, Li H, Yi X, Xiao C, Zhang D, Li M. Arf1 regulates the ER-mitochondria encounter structure (ERMES) in a reactive oxygen species-dependent manner. FEBS J 2018; 285:2004-2018. [PMID: 29603662 DOI: 10.1111/febs.14445] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 01/07/2018] [Accepted: 03/22/2018] [Indexed: 12/18/2022]
Abstract
The Arf family of small GTP-binding and -hydrolyzing proteins are some of the most important intracellular regulators of membrane dynamics. In this study, we identified the Golgi-localized Arf family G protein Arf1 in Candida albicans and confirmed its conserved function in regulating the secretory pathway. Interestingly, deletion of ARF1 resulted in intracellular reactive oxygen species (ROS) accumulation, and induced formation of the endoplasmic reticulum (ER)-mitochondria encounter structure (ERMES). Moreover, N-acetylcysteine-mediated ROS scavenging in the arf1Δ/Δ strain attenuated ERMES formation, indicating that intracellular ROS accumulation resulting from ARF1 deletion facilitated ERMES formation. In addition, Arf1 regulated many key physiological processes in C. albicans, including cell cycle progression, morphogenesis and virulence. This study uncovers a role for Arf family G proteins in regulating ERMES formation and sheds new light on the crucial contribution of ROS to membrane dynamics.
Collapse
Affiliation(s)
- Bing Zhang
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Qilin Yu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Da Huo
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Jianrong Li
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Chao Liang
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Hongyue Li
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Xiao Yi
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Chenpeng Xiao
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Dan Zhang
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Mingchun Li
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
36
|
Dunn MJ, Kinney GM, Washington PM, Berman J, Anderson MZ. Functional diversification accompanies gene family expansion of MED2 homologs in Candida albicans. PLoS Genet 2018; 14:e1007326. [PMID: 29630599 PMCID: PMC5908203 DOI: 10.1371/journal.pgen.1007326] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/19/2018] [Accepted: 03/21/2018] [Indexed: 01/03/2023] Open
Abstract
Gene duplication facilitates functional diversification and provides greater phenotypic flexibility to an organism. Expanded gene families arise through repeated gene duplication but the extent of functional divergence that accompanies each paralogous gene is generally unexplored because of the difficulty in isolating the effects of single family members. The telomere-associated (TLO) gene family is a remarkable example of gene family expansion, with 14 members in the more pathogenic Candida albicans relative to two TLO genes in the closely-related species C. dubliniensis. TLO genes encode interchangeable Med2 subunits of the major transcriptional regulatory complex Mediator. To identify biological functions associated with each C. albicans TLO, expression of individual family members was regulated using a Tet-ON system and the strains were assessed across a range of phenotypes involved in growth and virulence traits. All TLOs affected multiple phenotypes and a single phenotype was often affected by multiple TLOs, including simple phenotypes such as cell aggregation and complex phenotypes such as virulence in a Galleria mellonella model of infection. No phenotype was regulated by all TLOs, suggesting neofunctionalization or subfunctionalization of ancestral properties among different family members. Importantly, regulation of three phenotypes could be mapped to individual polymorphic sites among the TLO genes, including an indel correlated with two phenotypes, growth in sucrose and macrophage killing. Different selective pressures have operated on the TLO sequence, with the 5’ conserved Med2 domain experiencing purifying selection and the gene/clade-specific 3’ end undergoing extensive positive selection that may contribute to the impact of individual TLOs on phenotypic variability. Therefore, expansion of the TLO gene family has conferred unique regulatory properties to each paralog such that it influences a range of phenotypes. We posit that the genetic diversity associated with this expansion contributed to C. albicans success as a commensal and opportunistic pathogen. Gene duplication is a rapid mechanism to generate additional sequences for natural selection to act upon and confer greater organismal fitness. If additional copies of the gene are beneficial, this process may be repeated to produce an expanded gene family containing many copies of related sequences. Following duplication, individual gene family members may retain functions of the ancestral gene or acquire new functions through mutation. How functional diversification accompanies expansion into large gene families remains largely unexplored due to the difficulty in assessing individual genes in the presence of the remaining family members. Here, we addressed this question using an inducible promoter to regulate expression of individual genes of the TLO gene family in the commensal yeast and opportunistic pathogen Candida albicans, which encode components of a major transcriptional regulator. Induced expression of individual TLOs affected a wide range of phenotypes such that significant functional overlap occurred among TLO genes and most phenotypes were affected by more than one TLO. Induced expression of individual TLOs did not produce massive phenotypic effects in most cases, suggesting that functional overlap among TLO genes may buffer new mutations that arise. Specific sequence variants among the TLO genes correlated with certain phenotypes and these sequence variants did not necessarily correlate with sequence similarity across the entire gene. Therefore, individual TLO family members evolved specific functional roles following duplication that likely reflect a combination of inherited function and new mutation.
Collapse
Affiliation(s)
- Matthew J. Dunn
- Department of Microbiology, The Ohio State University, Columbus, OH, United States of America
| | - Griffin M. Kinney
- Department of Microbiology, The Ohio State University, Columbus, OH, United States of America
| | - Pamela M. Washington
- Department of Microbiology, The Ohio State University, Columbus, OH, United States of America
| | - Judith Berman
- Department of Molecular Microbiology and Biotechnology, Tel Aviv University, Tel Aviv, Israel
| | - Matthew Z. Anderson
- Department of Microbiology, The Ohio State University, Columbus, OH, United States of America
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States of America
- * E-mail:
| |
Collapse
|
37
|
Li C, Wang Y, Li Y, Yu Q, Jin X, Wang X, Jia A, Hu Y, Han L, Wang J, Yang H, Yan D, Bi Y, Liu G. HIF1α-dependent glycolysis promotes macrophage functional activities in protecting against bacterial and fungal infection. Sci Rep 2018; 8:3603. [PMID: 29483608 PMCID: PMC5827022 DOI: 10.1038/s41598-018-22039-9] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 02/15/2018] [Indexed: 02/06/2023] Open
Abstract
Macrophages are important innate immune defense system cells in the fight against bacterial and fungal pathogenic infections. They exhibit significant plasticity, particularly with their ability to undergo functional differentiation. Additionally, HIF1α is critically involved in the functional differentiation of macrophages during inflammation. However, the role of macrophage HIF1α in protecting against different pathogenic infections remains unclear. In this study, we investigated and compared the roles of HIF1α in different macrophage functional effects of bacterial and fungal infections in vitro and in vivo. We found that bacterial and fungal infections produced similar effects on macrophage functional differentiation. HIF1α deficiency inhibited pro-inflammatory macrophage functional activities when cells were stimulated with LPS or curdlan in vitro or when mice were infected with L. monocytogenes or C. albicans in vivo, thus decreasing pro-inflammatory TNFα and IL-6 secretion associated with pathogenic microorganism survival. Alteration of glycolytic pathway activation was required for the functional differentiation of pro-inflammatory macrophages in protecting against bacterial and fungal infections. Thus, the HIF1α-dependent glycolytic pathway is essential for pro-inflammatory macrophage functional differentiation in protecting against bacterial and fungal infections.
Collapse
Affiliation(s)
- Chunxiao Li
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, 100875, China
| | - Yu Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, 100875, China
| | - Yan Li
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, 100875, China
| | - Qing Yu
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, 100875, China
| | - Xi Jin
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, 100875, China
| | - Xiao Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, 100875, China
| | - Anna Jia
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, 100875, China
| | - Ying Hu
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, 100875, China
| | - Linian Han
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, 100875, China
| | - Jian Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, 100875, China
| | - Hui Yang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Dapeng Yan
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| | - Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China.
| | - Guangwei Liu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, 100875, China.
| |
Collapse
|
38
|
Rapala-Kozik M, Bochenska O, Zajac D, Karkowska-Kuleta J, Gogol M, Zawrotniak M, Kozik A. Extracellular proteinases of Candida species pathogenic yeasts. Mol Oral Microbiol 2018; 33:113-124. [PMID: 29139623 DOI: 10.1111/omi.12206] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2017] [Indexed: 02/06/2023]
Abstract
The increased incidence of severe disseminated infections caused by the opportunistic yeast-like fungi Candida spp. highlights the urgent need for research into the major virulence factors of these pathogens-extracellular aspartic proteinases of the candidapepsin and yapsin families. Classically, these enzymes were considered to be generally destructive factors that damage host tissues and provide nutrients for pathogen propagation. However, in recent decades, novel and more specific functions have been suggested for extracellular candidal proteinases. These include contributions to cell wall maintenance and remodeling, the formation of polymicrobial biofilms, adhesion to external protective barriers of the host, the deregulation of host proteolytic cascades (such as the complement system, blood coagulation and the kallikrein-kinin system), a dysregulated host proteinase-inhibitor balance, the inactivation of host antimicrobial peptides, evasion of immune responses and the induction of inflammatory mediator release from host cells. Only a few of these activities recognized in Candida albicans candidapepsins have been also confirmed in other Candida species, and characterization of Candida glabrata yapsins remains limited.
Collapse
Affiliation(s)
- M Rapala-Kozik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - O Bochenska
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - D Zajac
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - J Karkowska-Kuleta
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - M Gogol
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.,Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - M Zawrotniak
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - A Kozik
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
39
|
Ries LNA, Beattie S, Cramer RA, Goldman GH. Overview of carbon and nitrogen catabolite metabolism in the virulence of human pathogenic fungi. Mol Microbiol 2017; 107:277-297. [PMID: 29197127 DOI: 10.1111/mmi.13887] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 11/20/2017] [Accepted: 11/23/2017] [Indexed: 12/12/2022]
Abstract
It is estimated that fungal infections, caused most commonly by Candida albicans, Aspergillus fumigatus and Cryptococcus neoformans, result in more deaths annually than malaria or tuberculosis. It has long been hypothesized the fungal metabolism plays a critical role in virulence though specific nutrient sources utilized by human pathogenic fungi in vivo has remained enigmatic. However, the metabolic utilisation of preferred carbon and nitrogen sources, encountered in a host niche-dependent manner, is known as carbon catabolite and nitrogen catabolite repression (CCR, NCR), and has been shown to be important for virulence. Several sensory and uptake systems exist, including carbon and nitrogen source-specific sensors and transporters, that allow scavenging of preferred nutrient sources. Subsequent metabolic utilisation is governed by transcription factors, whose functions and essentiality differ between fungal species. Furthermore, additional factors exist that contribute to the implementation of CCR and NCR. The role of the CCR and NCR-related factors in virulence varies greatly between fungal species and a substantial gap in knowledge exists regarding specific pathways. Further elucidation of carbon and nitrogen metabolism mechanisms is therefore required in a fungal species- and animal model-specific manner in order to screen for targets that are potential candidates for anti-fungal drug development.
Collapse
Affiliation(s)
- Laure Nicolas Annick Ries
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes, Ribeirão Preto, São Paulo, 3900, CEP 14049-900, Brazil
| | - Sarah Beattie
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, 74 College Street Remsen 213, Hanover, NH 03755, USA
| | - Robert A Cramer
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, 74 College Street Remsen 213, Hanover, NH 03755, USA
| | - Gustavo H Goldman
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Avenida do Café s/n°, Ribeirão Preto, São Paulo, CEP 14040903, Brazil
| |
Collapse
|
40
|
Zhao Y, Tang H, Tan C, Zhao H, Liu Y. HLA-B27 Correlates with the Intracellular Elimination, Replication, and Trafficking of Salmonella Enteritidis Collected from Reactive Arthritis Patients. Med Sci Monit 2017; 23:5420-5429. [PMID: 29135969 PMCID: PMC5699175 DOI: 10.12659/msm.904681] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The aim of this study was to explore the correlation between HLA-B27 and the intracellular elimination, replication, and trafficking of Salmonella enteritidis (S. enteritidis) collected from patients with reactive arthritis. MATERIAL AND METHODS Confocal microscopy, flow cytometry, and sandwich enzyme-linked immunosorbent assay (ELISA) were employed in this study to evaluate the localization of proteins of interest, to assess the intracellular trafficking of S. enteritidis, and to measure the production of cytokines of interest. RESULTS HLA-B27 was negatively associated with intracellular S. enteritidis elimination in healthy human monocytes/macrophages. In S. enteritidis infected monocytes/macrophages, HLA-27B was also negatively correlated with bacteria elimination but positively related to bacteria replication. S. enteritidis did not co-localize with NRAMP1 and LAMP1/2 in HLA-B27 cells. S. enteritidis did not co-exist with transferrin or dextran within HLA-B27 and A2 cells. CONCLUSIONS HLA-B27 is closely associated with the intracellular elimination and replication of S. enteritidis. Replicated bacteria in HLA-B27 monocytic cells were located within unique vacuoles rather than disturbing host endocytosis.
Collapse
Affiliation(s)
- Yi Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China (mainland)
| | - Honghu Tang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China (mainland)
| | - Chunyu Tan
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China (mainland)
| | - Hua Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China (mainland)
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China (mainland)
| |
Collapse
|
41
|
Consideration of Invasive Fungal Infections in Immunocompetent Hosts. ARCHIVES OF CLINICAL INFECTIOUS DISEASES 2017. [DOI: 10.5812/archcid.66111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
42
|
de Sequeira DCM, Menezes RC, Oliveira MME, Antas PRZ, De Luca PM, de Oliveira-Ferreira J, Borba CDM. Experimental Hyalohyphomycosis by Purpureocillium lilacinum: Outcome of the Infection in C57BL/6 Murine Models. Front Microbiol 2017; 8:1617. [PMID: 28878763 PMCID: PMC5572354 DOI: 10.3389/fmicb.2017.01617] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 08/08/2017] [Indexed: 01/10/2023] Open
Abstract
Purpureocillium lilacinum is a filamentous, hyaline fungus considered an emerging pathogen in humans. The aim of our study was to evaluate the outcome of hyalohyphomycosis in C57BL/6 murine models inoculated with two clinical P. lilacinum isolates (S1 and S2). Each isolate was inoculated in mice randomly distributed in immunocompetent (CPT) and immunosuppressed (SPS) groups. Mice were evaluated at day 7, 21, and 45 after inoculation for histopathological analysis, recovery of fungal cells, and immunological studies. Histological analysis showed scarce conidia-like structures in lung tissue from CPT mice and a lot of fungal cells in SPS mice inoculated with S2 compared to mice inoculated with S1. The maximum recovery of fungal cells was seen in CPT mice inoculated with both isolates at day 7, but with mean significantly higher in those inoculated with S2 isolate. Phenotypical characterization of T cells showed TCD8+ lymphocytes predominance over TCD4+ in immunosuppressed mice infected and control groups. We also observed higher percentages of the central and effector memory/effector phenotype in CPT mice infected with S2 strain, especially in TCD8+ in the initial period of infection. Regulatory T cells showed higher percentages in immunosuppressed, predominantly after the acute phase. Our results showed that the P. lilacinum is a fungus capable to cause damages in competent and immunosuppressed experimental hosts. Furthermore, S2 isolate seems to cause more damage to the experimental host and it was possible to identify different cellular subsets involved in the mice immune response.
Collapse
Affiliation(s)
- Danielly C M de Sequeira
- Laboratory of Taxonomy, Biochemistry and Bioprospecting of Fungi, Oswaldo Cruz Institute, Oswaldo Cruz FoundationRio de Janeiro, Brazil.,Laboratory of Immunoparasitology, Oswaldo Cruz Institute, Oswaldo Cruz FoundationRio de Janeiro, Brazil
| | - Rodrigo C Menezes
- Laboratory of Clinical Research in Dermatozoonosis, Evandro Chagas National Institute of Infectology, Oswaldo Cruz FoundationRio de Janeiro, Brazil
| | - Manoel M E Oliveira
- Laboratory of Mycology, Evandro Chagas National Institute of Infectology, Oswaldo Cruz FoundationRio de Janeiro, Brazil
| | - Paulo R Z Antas
- Laboratory of Clinical Immunology, Oswaldo Cruz Institute, Oswaldo Cruz FoundationRio de Janeiro, Brazil
| | - Paula M De Luca
- Laboratory of Immunoparasitology, Oswaldo Cruz Institute, Oswaldo Cruz FoundationRio de Janeiro, Brazil
| | | | - Cintia de Moraes Borba
- Laboratory of Taxonomy, Biochemistry and Bioprospecting of Fungi, Oswaldo Cruz Institute, Oswaldo Cruz FoundationRio de Janeiro, Brazil
| |
Collapse
|
43
|
Halder LD, Abdelfatah MA, Jo EAH, Jacobsen ID, Westermann M, Beyersdorf N, Lorkowski S, Zipfel PF, Skerka C. Factor H Binds to Extracellular DNA Traps Released from Human Blood Monocytes in Response to Candida albicans. Front Immunol 2017; 7:671. [PMID: 28133459 PMCID: PMC5233719 DOI: 10.3389/fimmu.2016.00671] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 12/19/2016] [Indexed: 12/15/2022] Open
Abstract
Upon systemic infection with human pathogenic yeast Candida albicans (C. albicans), human monocytes and polymorph nuclear neutrophilic granulocytes are the first immune cells to respond and come into contact with C. albicans. Monocytes exert immediate candidacidal activity and inhibit germination, mediate phagocytosis, and kill fungal cells. Here, we show that human monocytes spontaneously respond to C. albicans cells via phagocytosis, decondensation of nuclear DNA, and release of this decondensed DNA in the form of extracellular traps (called monocytic extracellular traps: MoETs). Both subtypes of monocytes (CD14++CD16−/CD14+CD16+) formed MoETs within the first hours upon contact with C. albicans. MoETs were characterized by the presence of citrullinated histone, myeloperoxidase, lactoferrin, and elastase. MoETs were also formed in response to Staphylococcus aureus and Escherichia coli, indicating a general reaction of monocytes to infectious microbes. MoET induction differs from extracellular trap formation in macrophages as MoETs are not triggered by simvastatin, an inhibitor of cholesterol synthesis and inducer of extracellular traps in macrophages. Extracellular traps from both monocytes and neutrophils activate complement and C3b is deposited. However, factor H (FH) binds via C3b to the extracellular DNA, mediates cofactor activity, and inhibits the induction of the inflammatory cytokine interleukin-1 beta in monocytes. Altogether, the results show that human monocytes release extracellular DNA traps in response to C. albicans and that these traps finally bind FH via C3b to presumably support clearance without further inflammation.
Collapse
Affiliation(s)
- Luke D Halder
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology , Jena , Germany
| | - Mahmoud A Abdelfatah
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology , Jena , Germany
| | - Emeraldo A H Jo
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology , Jena , Germany
| | - Ilse D Jacobsen
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany; Friedrich-Schiller University, Jena, Germany
| | - Martin Westermann
- Center for Electron Microscopy of the University Hospital Jena , Jena , Germany
| | - Niklas Beyersdorf
- Institute for Virology and Immunobiology, University of Würzburg , Würzburg , Germany
| | - Stefan Lorkowski
- Institute of Nutrition, Friedrich-Schiller University , Jena , Germany
| | - Peter F Zipfel
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany; Friedrich-Schiller University, Jena, Germany
| | - Christine Skerka
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology , Jena , Germany
| |
Collapse
|
44
|
Poletto F, Lima F, Lundberg D, Nylander T, Loh W. Tailoring the internal structure of liquid crystalline nanoparticles responsive to fungal lipases: A potential platform for sustained drug release. Colloids Surf B Biointerfaces 2016; 147:210-216. [DOI: 10.1016/j.colsurfb.2016.08.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 05/27/2016] [Accepted: 08/02/2016] [Indexed: 01/04/2023]
|
45
|
Gabaldón T, Naranjo-Ortíz MA, Marcet-Houben M. Evolutionary genomics of yeast pathogens in the Saccharomycotina. FEMS Yeast Res 2016; 16:fow064. [PMID: 27493146 PMCID: PMC5815160 DOI: 10.1093/femsyr/fow064] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 05/18/2016] [Accepted: 08/02/2016] [Indexed: 02/06/2023] Open
Abstract
Saccharomycotina comprises a diverse group of yeasts that includes numerous species of industrial or clinical relevance. Opportunistic pathogens within this clade are often assigned to the genus Candida but belong to phylogenetically distant lineages that also comprise non-pathogenic species. This indicates that the ability to infect humans has evolved independently several times among Saccharomycotina. Although the mechanisms of infection of the main groups of Candida pathogens are starting to be unveiled, we still lack sufficient understanding of the evolutionary paths that led to a virulent phenotype in each of the pathogenic lineages. Deciphering what genomic changes underlie the evolutionary emergence of a virulence trait will not only aid the discovery of novel virulence mechanisms but it will also provide valuable information to understand how new pathogens emerge, and what clades may pose a future danger. Here we review recent comparative genomics efforts that have revealed possible evolutionary paths to pathogenesis in different lineages, focusing on the main three agents of candidiasis worldwide: Candida albicans, C. parapsilosis and C. glabrata We will discuss what genomic traits may facilitate the emergence of virulence, and focus on two different genome evolution mechanisms able to generate drastic phenotypic changes and which have been associated to the emergence of virulence: gene family expansion and interspecies hybridization.
Collapse
Affiliation(s)
- Toni Gabaldón
- Department of Bioinformatics and Genomics, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain Institució Catalana de Recerca i Estudis Avançats (ICREA), Pg. Lluís Companys 23, 08010 Barcelona, Spain
| | - Miguel A Naranjo-Ortíz
- Department of Bioinformatics and Genomics, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Marina Marcet-Houben
- Department of Bioinformatics and Genomics, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| |
Collapse
|
46
|
The Candida albicans fimbrin Sac6 regulates oxidative stress response (OSR) and morphogenesis at the transcriptional level. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2255-66. [DOI: 10.1016/j.bbamcr.2016.06.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 05/27/2016] [Accepted: 06/02/2016] [Indexed: 12/30/2022]
|
47
|
Chen Y, Yu Q, Wang H, Dong Y, Jia C, Zhang B, Xiao C, Zhang B, Xing L, Li M. The malfunction of peroxisome has an impact on the oxidative stress sensitivity in Candida albicans. Fungal Genet Biol 2016; 95:1-12. [PMID: 27473887 DOI: 10.1016/j.fgb.2016.07.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 07/24/2016] [Accepted: 07/26/2016] [Indexed: 11/28/2022]
Abstract
The peroxisome plays an essential role in eukaryotic cellular metabolism, including β-oxidation of fatty acids and detoxification of hydrogen peroxide. However, its functions in the important fungal pathogen, C. albicans, remain to be investigated. In this study, we identified a homologue of Saccharomyces cerevisiae peroxisomal protein Pex1 in this pathogen, and explored its functions in stress tolerance. Fluorescence observation revealed that C. albicans Pex1 was localized in the peroxisomes, and its loss led to the defect in peroxisome formation. Interestingly, the pex1Δ/Δ mutant had increased tolerance to oxidative stress, which was neither associated with the Cap1 pathway, nor related to the altered distribution of catalase. However, under oxidative stress, the pex1Δ/Δ mutant showed increased expression of autophagy-related genes, with enhanced cytoplasm-to-vacuole transport and degradation of the autophagy markers Atg8 and Lap41. Moreover, the double mutants pex1Δ/Δatg8Δ/Δ and pex1Δ/Δatg1Δ/Δ, both of which were defective in autophagy and peroxisome formation, showed remarkable attenuated tolerance to oxidative stress. These results indicated that autophagy is involved in resistance to oxidative stress in pex1Δ/Δ mutant. Taken together, this study provides evidence that the peroxisomal protein Pex1 regulates oxidative stress tolerance in an autophagy-dependent manner in C. albicans.
Collapse
Affiliation(s)
- Yulu Chen
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, PR China
| | - Qilin Yu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, PR China
| | - Honggang Wang
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, PR China
| | - Yijie Dong
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, PR China
| | - Chang Jia
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, PR China
| | - Bing Zhang
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, PR China
| | - Chenpeng Xiao
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, PR China
| | - Biao Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Laijun Xing
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, PR China
| | - Mingchun Li
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, PR China.
| |
Collapse
|
48
|
Brunke S, Mogavero S, Kasper L, Hube B. Virulence factors in fungal pathogens of man. Curr Opin Microbiol 2016; 32:89-95. [PMID: 27257746 DOI: 10.1016/j.mib.2016.05.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 04/25/2016] [Accepted: 05/17/2016] [Indexed: 01/29/2023]
Abstract
Human fungal pathogens are a commonly underestimated cause of severe diseases associated with high morbidity and mortality. Like other pathogens, their survival and growth in the host, as well as subsequent host damage, is thought to be mediated by virulence factors which set them apart from harmless microbes. In this review, we describe and discuss commonly employed strategies for fungal survival and growth in the host and how these affect the host-fungus interactions to lead to disease. While many of these strategies require host-specific virulence factors, more generally any fitness factor which allows growth under host-like conditions can be required for pathogenesis. Furthermore, we briefly summarize how different fungal pathogens are thought to damage the host. We find that in addition to a core of common activities relevant for growth, different groups of fungi employ different strategies which in spite of (or together with) the host's response can lead to disease.
Collapse
Affiliation(s)
- Sascha Brunke
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute Jena (HKI), Beutenbergstrasse 11a, 07745, Jena, Germany
| | - Selene Mogavero
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute Jena (HKI), Beutenbergstrasse 11a, 07745, Jena, Germany
| | - Lydia Kasper
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute Jena (HKI), Beutenbergstrasse 11a, 07745, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute Jena (HKI), Beutenbergstrasse 11a, 07745, Jena, Germany; Friedrich Schiller University, Jena, Germany; Center for Sepsis Control and Care (CSCC), Jena, Germany.
| |
Collapse
|
49
|
Miramón P, Lorenz MC. The SPS amino acid sensor mediates nutrient acquisition and immune evasion in Candida albicans. Cell Microbiol 2016; 18:1611-1624. [PMID: 27060451 DOI: 10.1111/cmi.12600] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 02/12/2016] [Accepted: 03/30/2016] [Indexed: 01/20/2023]
Abstract
Candida albicans is well adapted to its host and is able to sense and respond to the nutrients available within. We have shown that C. albicans avidly utilizes amino acids as a carbon source, which allows this opportunistic pathogen to neutralize acidic environments, including the macrophage phagosome. The transcription factor Stp2 is a key regulator of this phenomenon, and we sought to understand the mechanism of activation of Stp2, focusing on the SPS sensor system previously characterized for its role in nitrogen acquisition. We generated deletion mutants of the three components, SSY1, PTR3 and SSY5 and demonstrated that these strains utilize amino acids poorly as carbon source, cannot neutralize the medium in response to these nutrients, and have reduced ammonia release. Exogenous amino acids rapidly induce proteolytic processing of Stp2 and nuclear translocation in an SPS-dependent manner. A truncated version of Stp2, lacking the amino terminal nuclear exclusion domain, could suppress the growth and pH neutralization defects of the SPS mutants. We showed that the SPS system is required for normal resistance of C. albicans to macrophages and that mutants defective in this system reside in more acidic phagosomes compared with wild type cells; however, a more equivocal contribution was observed in the murine model of disseminated candidiasis. Taken together, these results indicate that the SPS system is activated under carbon starvation conditions resembling host environments, regulating Stp2 functions necessary for amino acid catabolism and normal interactions with innate immune cells.
Collapse
Affiliation(s)
- Pedro Miramón
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center, Houston, TX 77030, USA
| | - Michael C Lorenz
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center, Houston, TX 77030, USA.
| |
Collapse
|
50
|
The Endoplasmic Reticulum-Mitochondrion Tether ERMES Orchestrates Fungal Immune Evasion, Illuminating Inflammasome Responses to Hyphal Signals. mSphere 2016; 1:mSphere00074-16. [PMID: 27303738 PMCID: PMC4888881 DOI: 10.1128/msphere.00074-16] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 04/26/2016] [Indexed: 01/01/2023] Open
Abstract
The pathogenic yeast Candida albicans escapes macrophages by triggering NLRP3 inflammasome-dependent host cell death (pyroptosis). Pyroptosis is inflammatory and must be tightly regulated by host and microbe, but the mechanism is incompletely defined. We characterized the C. albicans endoplasmic reticulum (ER)-mitochondrion tether ERMES and show that the ERMES mmm1 mutant is severely crippled in killing macrophages despite hyphal formation and normal phagocytosis and survival. To understand dynamic inflammasome responses to Candida with high spatiotemporal resolution, we established live-cell imaging for parallel detection of inflammasome activation and pyroptosis at the single-cell level. This showed that the inflammasome response to mmm1 mutant hyphae is delayed by 10 h, after which an exacerbated activation occurs. The NLRP3 inhibitor MCC950 inhibited inflammasome activation and pyroptosis by C. albicans, including exacerbated inflammasome activation by the mmm1 mutant. At the cell biology level, inactivation of ERMES led to a rapid collapse of mitochondrial tubular morphology, slow growth and hyphal elongation at host temperature, and reduced exposed 1,3-β-glucan in hyphal populations. Our data suggest that inflammasome activation by C. albicans requires a signal threshold dependent on hyphal elongation and cell wall remodeling, which could fine-tune the response relative to the level of danger posed by C. albicans. The phenotypes of the ERMES mutant and the lack of conservation in animals suggest that ERMES is a promising antifungal drug target. Our data further indicate that NLRP3 inhibition by MCC950 could modulate C. albicans-induced inflammation. IMPORTANCE The yeast Candida albicans causes human infections that have mortality rates approaching 50%. The key to developing improved therapeutics is to understand the host-pathogen interface. A critical interaction is that with macrophages: intracellular Candida triggers the NLRP3/caspase-1 inflammasome for escape through lytic host cell death, but this also activates antifungal responses. To better understand how the inflammasome response to Candida is fine-tuned, we established live-cell imaging of inflammasome activation at single-cell resolution, coupled with analysis of the fungal ERMES complex, a mitochondrial regulator that lacks human homologs. We show that ERMES mediates Candida escape via inflammasome-dependent processes, and our data suggest that inflammasome activation is controlled by the level of hyphal growth and exposure of cell wall components as a proxy for severity of danger. Our study provides the most detailed dynamic analysis of inflammasome responses to a fungal pathogen so far and establishes promising pathogen- and host-derived therapeutic strategies.
Collapse
|