1
|
Bihaniya H, Rudraprasad D, Joseph J. Pathobiology of Fungal Endophthalmitis: A Major Review. ACS Infect Dis 2024; 10:3126-3137. [PMID: 39267469 DOI: 10.1021/acsinfecdis.4c00442] [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] [Indexed: 09/17/2024]
Abstract
Fungal endophthalmitis is a chronic inflammatory condition of the eye's posterior segment that can lead to irreversible vision loss. While relatively rare in western countries, its incidence is notably higher in Asia, particularly India. The condition's prevalence is exacerbated by factors such as intravenous drug use, antibiotics, and ocular surgeries. Fungal endophthalmitis can be categorized as endogenous, arising from systemic infection, or exogenous, linked to external sources such as trauma or surgery. The fungal agents responsible vary by region, with Candida species common in the West and Aspergillus and Fusarium species more prevalent in India. Management typically involves vitrectomy and intravitreal antifungal drugs such as amphotericin B and voriconazole, though treatment is often complicated by multidrug resistance and culture-negative cases. Recent proteomic and transcriptomic analyses have highlighted the early and sustained activation of the host immune response during infection involving key inflammatory and oxidative stress-related proteins. Given the potential for excessive inflammation to cause retinal damage, targeted immunotherapies are crucial. Immunomodulation, which aims to balance the immune response, shows promise in preserving vision while effectively combating the infection. Key targets for immunomodulation include pro-inflammatory cytokines (TNF-α, IL-1β, IL-6, IL-17), chemokines (CCL2, CXCL8), Toll-like receptors (TLR2, TLR4), and the complement system. Additionally, modulating the activity of macrophages, neutrophils, regulatory T cells, and Th17 cells, as well as targeting inflammasomes, can help control inflammation. Biologic agents and small molecule inhibitors offer further avenues for precise immune response modulation. This review underscores the importance of a comprehensive understanding of host-pathogen interactions in the development of effective therapies for fungal endophthalmitis.
Collapse
Affiliation(s)
- Himanshi Bihaniya
- Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad, Telangana 500034, India
| | - Dhanwini Rudraprasad
- Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad, Telangana 500034, India
- Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Joveeta Joseph
- Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad, Telangana 500034, India
| |
Collapse
|
2
|
Dühring S, Schuster S. Studying mixed-species biofilms of Candida albicans and Staphylococcus aureus using evolutionary game theory. PLoS One 2024; 19:e0297307. [PMID: 38446770 PMCID: PMC10917284 DOI: 10.1371/journal.pone.0297307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 01/03/2024] [Indexed: 03/08/2024] Open
Abstract
Mixed-species biofilms of Candida albicans and Staphylococcus aureus pose a significant clinical challenge due to their resistance to the human immune system and antimicrobial therapy. Using evolutionary game theory and nonlinear dynamics, we analyse the complex interactions between these organisms to understand their coexistence in the human host. We determine the Nash equilibria and evolutionary stable strategies of the game between C. albicans and S. aureus and point out different states of the mixed-species biofilm. Using replicator equations we study the fungal-bacterial interactions on a population level. Our focus is on the influence of available nutrients and the quorum sensing molecule farnesol, including the potential therapeutic use of artificially added farnesol. We also investigate the impact of the suggested scavenging of C. albicans hyphae by S. aureus. Contrary to common assumptions, we confirm the hypothesis that under certain conditions, mixed-species biofilms are not universally beneficial. Instead, different Nash equilibria occur depending on encountered conditions (i.e. varying farnesol levels, either produced by C. albicans or artificially added), including antagonism. We further show that the suggested scavenging of C. albicans' hyphae by S. aureus does not influence the overall outcome of the game. Moreover, artificially added farnesol strongly affects the dynamics of the game, although its use as a medical adjuvant (add-on medication) may pose challenges.
Collapse
Affiliation(s)
- Sybille Dühring
- Department of Bioinformatics, Friedrich-Schiller-University Jena, Jena, Germany
| | - Stefan Schuster
- Department of Bioinformatics, Friedrich-Schiller-University Jena, Jena, Germany
| |
Collapse
|
3
|
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
|
4
|
Daud M, Dasari P, Adelfinger M, Langenhorst D, Lother J, Slavkovic-Lukic D, Berges C, Kruhm M, Galler A, Schleussner C, Luther CH, Alberter K, Althammer A, Shaikh H, Pallmann N, Bodem J, El-Mowafy M, Beilhack A, Dittrich M, Topp MS, Zipfel PF, Beyersdorf N. Enolase 1 of Candida albicans binds human CD4 + T cells and modulates naïve and memory responses. Eur J Immunol 2023; 53:e2250284. [PMID: 37503840 DOI: 10.1002/eji.202250284] [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: 11/23/2022] [Revised: 06/05/2023] [Accepted: 07/27/2023] [Indexed: 07/29/2023]
Abstract
To obtain a better understanding of the biology behind life-threatening fungal infections caused by Candida albicans, we recently conducted an in silico screening for fungal and host protein interaction partners. We report here that the extracellular domain of human CD4 binds to the moonlighting protein enolase 1 (Eno1) of C. albicans as predicted bioinformatically. By using different anti-CD4 monoclonal antibodies, we determined that C. albicans Eno1 (CaEno1) primarily binds to the extracellular domain 3 of CD4. Functionally, we observed that CaEno1 binding to CD4 activated lymphocyte-specific protein tyrosine kinase (LCK), which was also the case for anti-CD4 monoclonal antibodies tested in parallel. CaEno1 binding to naïve human CD4+ T cells skewed cytokine secretion toward a Th2 profile indicative of poor fungal control. Moreover, CaEno1 inhibited human memory CD4+ T-cell recall responses. Therapeutically, CD4+ T cells transduced with a p41/Crf1-specific T-cell receptor developed for adoptive T-cell therapy were not inhibited by CaEno1 in vitro. Together, the interaction of human CD4+ T cells with CaEno1 modulated host CD4+ T-cell responses in favor of the fungus. Thus, CaEno1 mediates not only immune evasion through its interference with complement regulators but also through the direct modulation of CD4+ T-cell responses.
Collapse
Affiliation(s)
- Muhammad Daud
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Prasad Dasari
- Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany
| | - Marion Adelfinger
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | - Daniela Langenhorst
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Jasmin Lother
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | - Dragana Slavkovic-Lukic
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | - Carsten Berges
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | - Michaela Kruhm
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | | | | | | | - Karl Alberter
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Anton Althammer
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Haroon Shaikh
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | - Niklas Pallmann
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Jochen Bodem
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Mohammed El-Mowafy
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
- Faculty of Pharmacy, Department of Microbiology & Immunology, Mansoura University, Mansoura, Egypt
| | - Andreas Beilhack
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | - Marcus Dittrich
- Chair of Bioinformatics, University of Würzburg, Würzburg, Germany
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Max S Topp
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | - Peter F Zipfel
- Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany
- Friedrich Schiller University, Jena, Germany
| | - Niklas Beyersdorf
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
5
|
Langenhorst D, Fürst AL, Alberter K, Vilhena C, Dasari P, Daud M, Heilig L, Luther CH, Dittrich M, Reiher N, Wich M, Elmowafy M, Jacobsen ID, Jungnickel B, Zipfel PF, Beyersdorf N. Soluble Enolase 1 of Candida albicans and Aspergillus fumigatus Stimulates Human and Mouse B Cells and Monocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:804-815. [PMID: 37436030 DOI: 10.4049/jimmunol.2200318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/23/2023] [Indexed: 07/13/2023]
Abstract
Because of the growing numbers of immunocompromised patients, the incidence of life-threatening fungal infections caused by Candida albicans and Aspergillus fumigatus is increasing. We have recently identified enolase 1 (Eno1) from A. fumigatus as an immune evasion protein. Eno1 is a fungal moonlighting protein that mediates adhesion and invasion of human cells and also immune evasion through complement inactivation. We now show that soluble Eno1 has immunostimulatory activity. We observed that Eno1 from both C. albicans and A. fumigatus directly binds to the surface of lymphocytes, preferentially human and mouse B cells. Functionally, Eno1 upregulated CD86 expression on B cells and induced proliferation. Although the receptor for fungal Eno1 on B lymphocytes is still unknown, the comparison of B cells from wild-type and MyD88-deficient mice showed that B cell activation by Eno1 required MyD88 signaling. With respect to infection biology, we noted that mouse B cells stimulated by Eno1 secreted IgM and IgG2b. These Igs bound C. albicans hyphae in vitro, suggesting that Eno1-induced Ab secretion might contribute to protection from invasive fungal disease in vivo. Eno1 also triggered the release of proinflammatory cytokines from monocytes, particularly IL-6, which is a potent activator of B cells. Together, our data shed new light on the role of secreted Eno1 in infections with C. albicans and A. fumigatus. Eno1 secretion by these pathogenic microbes appears to be a double-edged sword by supporting fungal pathogenicity while triggering (antifungal) immunity.
Collapse
Affiliation(s)
- Daniela Langenhorst
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Anna-Lisa Fürst
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Karl Alberter
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Cláudia Vilhena
- Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany
| | - Prasad Dasari
- Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany
| | - Muhammad Daud
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Linda Heilig
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | | | - Marcus Dittrich
- Chair of Bioinformatics, University of Würzburg, Würzburg, Germany
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Nadine Reiher
- Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany
| | | | - Mohammed Elmowafy
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
- Department of Microbiology & Immunology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Ilse D Jacobsen
- Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany
- Friedrich Schiller University, Jena, Germany
| | | | - Peter F Zipfel
- Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany
- Friedrich Schiller University, Jena, Germany
| | - Niklas Beyersdorf
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
6
|
Kumar D, Kumar A. Cellular Attributes of Candida albicans Biofilm-Associated in Resistance Against Multidrug and Host Immune System. Microb Drug Resist 2023; 29:423-437. [PMID: 37428599 DOI: 10.1089/mdr.2022.0347] [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] [Indexed: 07/12/2023] Open
Abstract
One of the ubiquitous hospital-acquired infections is associated with Candida albicans fungus. Usually, this commensal fungus causes no harm to its human host, as it lives mutually with mucosal/epithelial tissue surface cells. Nevertheless, due to the activity of various immune weakening factors, this commensal starts reinforcing its virulence attributes with filamentation/hyphal growth and building an absolute microcolony composed of yeast, hyphal, and pseudohyphal cells, which is suspended in an extracellular gel-like polymeric substance (EPS) called biofilms. This polymeric substance is the mixture of the secreted compounds from C. albicans as well as several host cell proteins. Indeed, the presence of these host factors makes their identification and differentiation process difficult by host immune components. The gel-like texture of the EPS makes it sticky, which adsorbs most of the extracolonial compounds traversing through it that aid in penetration hindrance. All these factors further contribute to the multidrug resistance phenotype of C. albicans biofilm that is spotlighted in this article. The mechanisms it employs to escape the host immune system are also addressed effectively. The article focuses on cellular and molecular determinants involved in the resistance of C. albicans biofilm against multidrug and the host immune system.
Collapse
Affiliation(s)
- Dushyant Kumar
- Department of Biotechnology, National Institute of Technology, Raipur, India
| | - Awanish Kumar
- Department of Biotechnology, National Institute of Technology, Raipur, India
| |
Collapse
|
7
|
Dandekar T, Kunz M. Bioinformatics Connects Life with the Universe and All the Rest. Bioinformatics 2023. [DOI: 10.1007/978-3-662-65036-3_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
|
8
|
Melese AS. MODELLING OF PATHOGENS IMPACT ON THE HUMAN DISEASE TRANSMISSION WITH OPTIMAL CONTROL STRATEGIES. JOURNAL OF MATHEMATICAL SCIENCES 2022. [PMCID: PMC9628469 DOI: 10.1007/s10958-022-06027-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
This study concentrates on a nonlinear deterministic mathematical model for the impact of pathogens on human disease transmission with optimal control strategies. Both pathogen-free and coexistence equilibria are computed. The basic reproduction number R0, which plays a vital role in mathematical epidemiology, was derived. The qualitative analysis of the model revealed the scenario for both pathogen-free and coexistence equilibria together with R0. The local stability of the equilibria is established via the Jacobian matrix and Routh-Hurwitz criteria, while the global stability of the equilibria is proven by using an appropriate Lyapunov function. Also, the normalized sensitivity analysis has been performed to observe the impact of different parameters on R0. The proposed model is extended into optimal control problem by incorporating three control variables, namely, preventive measure variable based on separation of susceptible from contacting the pathogens, integrated vector management based on chemical, biological control, ... etc. to kill pathogens and their carriers, and supporting infective medication variable based on the care of the infected individual in quarantine center. Optimal disease control analysis is examined using Pontryagin minimum principle. Numerical simulations are performed depending on analytical results and discussed quantitatively.
Collapse
Affiliation(s)
- Abdisa Shiferaw Melese
- Department of Applied Mathematics, Adama Science and Technology University, Adama, Ethiopia
| |
Collapse
|
9
|
Holzknecht J, Dubrac S, Hedtrich S, Galgóczy L, Marx F. Small, Cationic Antifungal Proteins from Filamentous Fungi Inhibit Candida albicans Growth in 3D Skin Infection Models. Microbiol Spectr 2022; 10:e0029922. [PMID: 35499318 PMCID: PMC9241769 DOI: 10.1128/spectrum.00299-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/01/2022] [Indexed: 12/19/2022] Open
Abstract
The emerging resistance of human-pathogenic fungi to antifungal drugs urges the development of alternative therapeutic strategies. The small, cationic antifungal proteins (AFPs) from filamentous ascomycetes represent promising candidates for next-generation antifungals. These bio-molecules need to be tested for tolerance in the host and efficacy against fungal pathogens before they can be safely applied in humans. Testing of the efficacy and possible adverse effects of new drug candidates in three-dimensional (3D) human-cell based models represents an advantageous alternative to animal experiments. In, this study, as a proof-of-principle, we demonstrate the usefulness of 3D skin infection models for screening new antifungal drug candidates for topical application. We established a cutaneous infection with the opportunistic human-pathogenic yeast Candida albicans in a commercially available 3D full-thickness (FT) skin model to test the curative potential of distinct AFPs from Penicillium chrysogenum (PAFopt, PAFB, and PAFC) and Neosartorya (Aspergillus) fischeri (NFAP2) in vitro. All tested AFPs were comparably well tolerated by the skin models. The infected 3D models exhibited reduced epidermal permeability barriers, allowing C. albicans to colonize the epidermal and dermal layers, and showed increased secretion of the pro-inflammatory cytokine IL-6 and the chemokine IL-8. AFP treatment diminished the fungal burden and penetration depth of C. albicans in the infected models. The epidermal permeability barrier was restored and the secretion of IL-8 was decreased following AFP treatment. In summary, our study proves that the tested AFPs exhibit antifungal potential against cutaneous C. albicans infection in a 3D FT skin model. IMPORTANCE Candida albicans represents one of the most prevalent opportunistic fungal pathogens, causing superficial skin and mucosal infections in humans with certain predisposing health conditions and life-threatening systemic infections in immunosuppressed patients. The emerging drug resistance of this human-pathogenic yeast and the limited number of antifungal drugs for prevention and treatment of infections urgently demands the identification of new antifungal compounds with novel mechanisms of action. Small, cationic antifungal proteins (AFPs) from filamentous fungi represent promising candidates for next-generation antifungals for topical application. These bio-molecules need to be tested for tolerance by the host and efficacy in pathogen clearance prior to being involved in clinical trials. In a proof-of-principle study, we provide evidence for the suitability of 3D human-cell based models as advantageous alternatives to animal experiments. We document the tolerance of specific AFPs and their curative efficacy against cutaneous C. albicans infection in a 3D skin model.
Collapse
Affiliation(s)
- Jeanett Holzknecht
- Biocenter, Institute of Molecular Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sandrine Dubrac
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sarah Hedtrich
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - László Galgóczy
- Department of Biotechnology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
- Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
| | - Florentine Marx
- Biocenter, Institute of Molecular Biology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
10
|
Zheng Q, Duan L, Zhang Y, Li J, Zhang S, Wang H. A dynamically evolving war between autophagy and pathogenic microorganisms. J Zhejiang Univ Sci B 2022; 23:19-41. [PMID: 35029086 PMCID: PMC8758936 DOI: 10.1631/jzus.b2100285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Autophagy is an intracellular degradation process that maintains cellular homeostasis. It is essential for protecting organisms from environmental stress. Autophagy can help the host to eliminate invading pathogens, including bacteria, viruses, fungi, and parasites. However, pathogens have evolved multiple strategies to interfere with autophagic signaling pathways or inhibit the fusion of autophagosomes with lysosomes to form autolysosomes. Moreover, host cell matrix degradation by different types of autophagy can be used for the proliferation and reproduction of pathogens. Thus, determining the roles and mechanisms of autophagy during pathogen infections will promote understanding of the mechanisms of pathogen‒host interactions and provide new strategies for the treatment of infectious diseases.
Collapse
Affiliation(s)
- Qianqian Zheng
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China
| | - Liangwei Duan
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China
| | - Yang Zhang
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China
| | - Jiaoyang Li
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China
| | - Shiyu Zhang
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China
| | - Hui Wang
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China. .,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China.
| |
Collapse
|
11
|
Hady MA, Darwish AB, Abdel-Aziz MS, Sayed OM. Design of transfersomal nanocarriers of nystatin for combating vulvovaginal candidiasis; A different prospective. Colloids Surf B Biointerfaces 2021; 211:112304. [PMID: 34959094 DOI: 10.1016/j.colsurfb.2021.112304] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 11/26/2021] [Accepted: 12/20/2021] [Indexed: 11/25/2022]
Abstract
The objective of this study was to prepare and evaluate Nystatin (NYS) loaded transfersomes to achieve better treatment of vulvovaginal candidiasis. Nystatin transferosomes were formulated utilizing thin film hydration method. A 32 full factorial design was employed to evaluate the effect of different formulation variables. Two independent variables were chosen; the ratio between lecithin surfactant (X1) was set at three levels (10-40), and the type of surfactants (X2) was set at three levels (Span 60, Span 85 and Pluronic F-127). The dependent responses were; entrapment efficiency (Y1: EE %), vesicles size (Y2: VS) and release rate (Y3: RR). Design Expert® software was utilized to statistically optimize formulation variables. The vesicles revealed high NYS encapsulation efficiency ranging from 97.35 ± 0.03 to 98.01 ± 0.20% whereas vesicle size ranged from 194.8 ± 20.42 to 400.8 ± 42.09 nm. High negative zeta potential values indicated good stability of the prepared formulations. NYS release from transfersomes was biphasic and the release pattern followed Higuchi's model. The optimized formulation (F7) exhibited spherical morphology under transmission electron microscopy (TEM). In-vitro and in-vivo antifungal efficiency studies revealed that the optimized formula F7 exhibited significant eradication of candida infestation in comparison to free NYS. The results revealed that the developed NYS transfersomes could be a promising drug delivery system to enhance antifungal efficacy of NYS.
Collapse
Affiliation(s)
- Mayssa Abdel Hady
- Department of Pharmaceutical Technology, National Research Centre, El Bohouth Street, Cairo12622, Egypt
| | - Asmaa B Darwish
- Department of Pharmaceutical Technology, National Research Centre, El Bohouth Street, Cairo12622, Egypt.
| | - Mohamed S Abdel-Aziz
- Microbial Chemistry Department, Genetic Engineering and Biotechnology Division, National Research Centre, El Bohouth Street, Cairo 12622, Egypt
| | - Ossama M Sayed
- Department of Pharmaceutics Industrial Pharmacy, Faculty of Pharmacy, Sinai University - Kantara Branch, Egypt.
| |
Collapse
|
12
|
A. L. Bataineh MT, Soares NC, Semreen MH, Cacciatore S, Dash NR, Hamad M, Mousa MK, Salam JSA, Al Gharaibeh MF, Zerbini LF, Hamad M. Candida albicans PPG1, a serine/threonine phosphatase, plays a vital role in central carbon metabolisms under filament-inducing conditions: A multi-omics approach. PLoS One 2021; 16:e0259588. [PMID: 34874940 PMCID: PMC8651141 DOI: 10.1371/journal.pone.0259588] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/21/2021] [Indexed: 01/01/2023] Open
Abstract
Candida albicans is the leading cause of life-threatening bloodstream candidiasis, especially among immunocompromised patients. The reversible morphological transition from yeast to hyphal filaments in response to host environmental cues facilitates C. albicans tissue invasion, immune evasion, and dissemination. Hence, it is widely considered that filamentation represents one of the major virulence properties in C. albicans. We have previously characterized Ppg1, a PP2A-type protein phosphatase that controls filament extension and virulence in C. albicans. This study conducted RNA sequencing analysis of samples obtained from C. albicans wild type and ppg1Δ/Δ strains grown under filament-inducing conditions. Overall, ppg1Δ/Δ strain showed 1448 upregulated and 710 downregulated genes, representing approximately one-third of the entire annotated C. albicans genome. Transcriptomic analysis identified significant downregulation of well-characterized genes linked to filamentation and virulence, such as ALS3, HWP1, ECE1, and RBT1. Expression analysis showed that essential genes involved in C. albicans central carbon metabolisms, including GDH3, GPD1, GPD2, RHR2, INO1, AAH1, and MET14 were among the top upregulated genes. Subsequent metabolomics analysis of C. albicans ppg1Δ/Δ strain revealed a negative enrichment of metabolites with carboxylic acid substituents and a positive enrichment of metabolites with pyranose substituents. Altogether, Ppg1 in vitro analysis revealed a link between metabolites substituents and filament formation controlled by a phosphatase to regulate morphogenesis and virulence.
Collapse
Affiliation(s)
- Mohammad Tahseen A. L. Bataineh
- College of Medicine, University of Sharjah, Sharjah, UAE
- Research Institute for Medical & Health Sciences at University of Sharjah, Sharjah, UAE
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, UAE
- Department of Genetics and Molecular Biology, College Of Medicine And Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, UAE
| | - Nelson Cruz Soares
- Research Institute for Medical & Health Sciences at University of Sharjah, Sharjah, UAE
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, UAE
| | - Mohammad Harb Semreen
- Research Institute for Medical & Health Sciences at University of Sharjah, Sharjah, UAE
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, UAE
| | - Stefano Cacciatore
- Cancer Genomics Group, International Centre for Genetic Engineering and Biotechnology, Cape Town, South Africa
- Institute for Reproductive and Developmental Biology, Imperial College, London, United Kingdom
| | | | - Mohamad Hamad
- Research Institute for Medical & Health Sciences at University of Sharjah, Sharjah, UAE
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah, UAE
| | - Muath Khairi Mousa
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, UAE
| | | | | | - Luiz F. Zerbini
- Cancer Genomics Group, International Centre for Genetic Engineering and Biotechnology, Cape Town, South Africa
| | - Mawieh Hamad
- Research Institute for Medical & Health Sciences at University of Sharjah, Sharjah, UAE
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah, UAE
| |
Collapse
|
13
|
Radionuclide Imaging of Invasive Fungal Disease in Immunocompromised Hosts. Diagnostics (Basel) 2021; 11:diagnostics11112057. [PMID: 34829403 PMCID: PMC8620393 DOI: 10.3390/diagnostics11112057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 12/19/2022] Open
Abstract
Invasive fungal disease (IFD) leads to increased mortality, morbidity, and costs of treatment in patients with immunosuppressive conditions. The definitive diagnosis of IFD relies on the isolation of the causative fungal agents through microscopy, culture, or nucleic acid testing in tissue samples obtained from the sites of the disease. Biopsy is not always feasible or safe to be undertaken in immunocompromised hosts at risk of IFD. Noninvasive diagnostic techniques are, therefore, needed for the diagnosis and treatment response assessment of IFD. The available techniques that identify fungal-specific antigens in biological samples for diagnosing IFD have variable sensitivity and specificity. They also have limited utility in response assessment. Imaging has, therefore, been applied for the noninvasive detection of IFD. Morphologic imaging with computed tomography (CT) and magnetic resonance imaging (MRI) is the most applied technique. These techniques are neither sufficiently sensitive nor specific for the early diagnosis of IFD. Morphologic changes evaluated by CT and MRI occur later in the disease course and during recovery after successful treatment. These modalities may, therefore, not be ideal for early diagnosis and early response to therapy determination. Radionuclide imaging allows for targeting the host response to pathogenic fungi or specific structures of the pathogen itself. This makes radionuclide imaging techniques suitable for the early diagnosis and treatment response assessment of IFD. In this review, we aimed to discuss the interplay of host immunity, immunosuppression, and the occurrence of IFD. We also discuss the currently available radionuclide probes that have been evaluated in preclinical and clinical studies for their ability to detect IFD.
Collapse
|
14
|
Varahan S, Laxman S. Bend or break: how biochemically versatile molecules enable metabolic division of labor in clonal microbial communities. Genetics 2021; 219:iyab109. [PMID: 34849891 PMCID: PMC8633146 DOI: 10.1093/genetics/iyab109] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 06/29/2021] [Indexed: 02/05/2023] Open
Abstract
In fluctuating nutrient environments, isogenic microbial cells transition into "multicellular" communities composed of phenotypically heterogeneous cells, showing functional specialization. In fungi (such as budding yeast), phenotypic heterogeneity is often described in the context of cells switching between different morphotypes (e.g., yeast to hyphae/pseudohyphae or white/opaque transitions in Candida albicans). However, more fundamental forms of metabolic heterogeneity are seen in clonal Saccharomyces cerevisiae communities growing in nutrient-limited conditions. Cells within such communities exhibit contrasting, specialized metabolic states, and are arranged in distinct, spatially organized groups. In this study, we explain how such an organization can stem from self-organizing biochemical reactions that depend on special metabolites. These metabolites exhibit plasticity in function, wherein the same metabolites are metabolized and utilized for distinct purposes by different cells. This in turn allows cell groups to function as specialized, interdependent cross-feeding systems which support distinct metabolic processes. Exemplifying a system where cells exhibit either gluconeogenic or glycolytic states, we highlight how available metabolites can drive favored biochemical pathways to produce new, limiting resources. These new resources can themselves be consumed or utilized distinctly by cells in different metabolic states. This thereby enables cell groups to sustain contrasting, even apparently impossible metabolic states with stable transcriptional and metabolic signatures for a given environment, and divide labor in order to increase community fitness or survival. We speculate on possible evolutionary implications of such metabolic specialization and division of labor in isogenic microbial communities.
Collapse
Affiliation(s)
- Sriram Varahan
- Institute for Stem Cell Science and Regenerative Medicine (inStem), Bengaluru 560065, India
| | - Sunil Laxman
- Institute for Stem Cell Science and Regenerative Medicine (inStem), Bengaluru 560065, India
| |
Collapse
|
15
|
Bahattab O, Khan I, Bawazeer S, Rauf A, Qureshi MN, Al-Awthan YS, Muhammad N, Khan A, Akram M, Islam MN, Bin Emran T. Synthesis and biological activities of alcohol extract of black cumin seeds ( Bunium persicum)-based gold nanoparticles and their catalytic applications. GREEN PROCESSING AND SYNTHESIS 2021; 10:440-455. [DOI: 10.1515/gps-2021-0041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Abstract
Fast, simple, and environmentally friendly gold nanoparticles (Au-NPs) capped and stabilized with black cumin (Bunium persicum) seed alcohol extract are reported. The aqueous gold ions (Au3+) were treated with B. persicum (BP) seed extract, which resulted in a rapid color change to red, indicating the synthesis of Au-NPs. UV-Vis spectroscopy, FTIR, SEM, energy dispersive X-ray (EDX), and X-ray diffraction (XRD) techniques were used to further characterize the Au-NPs. Its stability was assessed against various pH levels and sodium chloride levels (NaCl), different salts of same concentration as well as at a range of temperature (30–100°C). The UV-Vis spectrum in the Au-NPs produced a 540 nm plasmon surface resonance, and a 25–50 nm range of particulates was shown in the SEM analysis. In addition, the FTIR spectra confirmed the inclusion in the capping and decrease of Au-NPs of amines, amide groups, and alcohols. The EDX analysis confirmed the presence of element Au. Furthermore, Au-NPs were tested for enzyme inhibition and antibacterial and antifungal activities and showed remarkable response. These findings have concluded that BP seed extract is an effective bio-reductant of gold nanoparticle synthesis, which can be further applied in different biomedical and pharmaceutical industries.
Collapse
Affiliation(s)
- Omar Bahattab
- Department of Biology, Faculty of Science, University of Tabuk , Tabuk , Saudi Arabia
| | - Ibrahim Khan
- Department of Chemistry, University of Swabi , Swabi , Anbar, KPK , Pakistan
| | - Sami Bawazeer
- Pharmacognosy Department, College of Pharmacy, Umm Al-Qura University , Makkah , Saudi Arabia
| | - Abdur Rauf
- Department of Chemistry, University of Swabi , Swabi , Anbar, KPK , Pakistan
| | | | - Yahya S. Al-Awthan
- Department of Biology, Faculty of Science, University of Tabuk , Tabuk , Saudi Arabia
- Department of Biology, Faculty of Science, Ibb University , Ibb , Yemen
| | - Naveed Muhammad
- Department of Pharmacy, Abdul Wali Khan University , Malakand , KPK , Pakistan
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa , P.O. Box 33, Birkat Al Mauz 616 , Nizwa , Oman
| | - Muhammad Akram
- Government Post Graduate College Charsadda , Charsadda , KPK , Pakistan
| | - Mohammad Nazmul Islam
- Department of Pharmacy, International Islamic University Chittagong , Chittagong , 4318 , Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh , Chittagong , 4381 , Bangladesh
| |
Collapse
|
16
|
Jenull S, Mair T, Tscherner M, Penninger P, Zwolanek F, Silao FGS, de San Vicente KM, Riedelberger M, Bandari NC, Shivarathri R, Petryshyn A, Chauhan N, Zacchi LF, -Landmann SL, Ljungdahl PO, Kuchler K. The histone chaperone HIR maintains chromatin states to control nitrogen assimilation and fungal virulence. Cell Rep 2021; 36:109406. [PMID: 34289370 PMCID: PMC8493472 DOI: 10.1016/j.celrep.2021.109406] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 03/10/2021] [Accepted: 06/24/2021] [Indexed: 12/13/2022] Open
Abstract
Adaptation to changing environments and immune evasion is pivotal for fitness of pathogens. Yet, the underlying mechanisms remain largely unknown. Adaptation is governed by dynamic transcriptional re-programming, which is tightly connected to chromatin architecture. Here, we report a pivotal role for the HIR histone chaperone complex in modulating virulence of the human fungal pathogen Candida albicans. Genetic ablation of HIR function alters chromatin accessibility linked to aberrant transcriptional responses to protein as nitrogen source. This accelerates metabolic adaptation and increases the release of extracellular proteases, which enables scavenging of alternative nitrogen sources. Furthermore, HIR controls fungal virulence, as HIR1 deletion leads to differential recognition by immune cells and hypervirulence in a mouse model of systemic infection. This work provides mechanistic insights into chromatin-coupled regulatory mechanisms that fine-tune pathogen gene expression and virulence. Furthermore, the data point toward the requirement of refined screening approaches to exploit chromatin modifications as antifungal strategies.
Collapse
Affiliation(s)
- Sabrina Jenull
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, 1030 Vienna, Austria
| | - Theresia Mair
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, 1030 Vienna, Austria
| | - Michael Tscherner
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, 1030 Vienna, Austria
| | - Philipp Penninger
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, 1030 Vienna, Austria
| | - Florian Zwolanek
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, 1030 Vienna, Austria
| | - Fitz-Gerald S Silao
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91 Stockholm, Sweden
| | - Kontxi Martinez de San Vicente
- Section of Immunology, Vetsuisse Faculty, University of Zürich, 8006 Zürich, Switzerland; Institute of Experimental Immunology, University of Zürich, 8057 Zürich, Switzerland
| | - Michael Riedelberger
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, 1030 Vienna, Austria
| | - Naga C Bandari
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Raju Shivarathri
- Public Health Research Institute, Rutgers, The State University of New Jersey, Newark, NJ 07102, USA
| | - Andriy Petryshyn
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, 1030 Vienna, Austria
| | - Neeraj Chauhan
- Public Health Research Institute, Rutgers, The State University of New Jersey, Newark, NJ 07102, USA; Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Lucia F Zacchi
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Salomé LeibundGut -Landmann
- Section of Immunology, Vetsuisse Faculty, University of Zürich, 8006 Zürich, Switzerland; Institute of Experimental Immunology, University of Zürich, 8057 Zürich, Switzerland
| | - Per O Ljungdahl
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91 Stockholm, Sweden
| | - Karl Kuchler
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, 1030 Vienna, Austria.
| |
Collapse
|
17
|
Abstract
Albumin is abundant in serum but is also excreted at mucosal surfaces and enters tissues when inflammation increases vascular permeability. Host-associated opportunistic pathogens encounter albumin during commensalism and when causing infections. Considering the ubiquitous presence of albumin, we investigated its role in the pathogenesis of infections with the model human fungal pathogen, Candida albicans. Albumin was introduced in various in vitro models that mimic different stages of systemic or mucosal candidiasis, where it reduced the ability of C. albicans to damage host cells. The amphipathic toxin candidalysin mediates necrotic host cell damage induced by C. albicans. Using cellular and biophysical assays, we determined that albumin functions by neutralizing candidalysin through hydrophobic interactions. We discovered that albumin, similarly, can neutralize a variety of fungal (α-amanitin), bacterial (streptolysin O and staurosporin), and insect (melittin) hydrophobic toxins. These data suggest albumin as a defense mechanism against toxins, which can play a role in the pathogenesis of microbial infections. IMPORTANCE Albumin is the most abundant serum protein in humans. During inflammation, serum albumin levels decrease drastically, and low albumin levels are associated with poor patient outcome. Thus, albumin may have specific functions during infection. Here, we describe the ability of albumin to neutralize hydrophobic microbial toxins. We show that albumin can protect against damage induced by the pathogenic yeast C. albicans by neutralizing its cytolytic toxin candidalysin. These findings suggest that albumin is a toxin-neutralizing protein that may play a role during infections with toxin-producing microorganisms.
Collapse
|
18
|
Zhou Y, Cheng L, Lei YL, Ren B, Zhou X. The Interactions Between Candida albicans and Mucosal Immunity. Front Microbiol 2021; 12:652725. [PMID: 34234752 PMCID: PMC8255368 DOI: 10.3389/fmicb.2021.652725] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 05/31/2021] [Indexed: 02/05/2023] Open
Abstract
Mucosa protects the body against external pathogen invasion. However, pathogen colonies on the mucosa can invade the mucosa when the immunosurveillance is compromised, causing mucosal infection and subsequent diseases. Therefore, it is necessary to timely and effectively monitor and control pathogenic microorganisms through mucosal immunity. Candida albicans is the most prevalent fungi on the mucosa. The C. albicans colonies proliferate and increase their virulence, causing severe infectious diseases and even death, especially in immunocompromised patients. The normal host mucosal immune defense inhibits pathogenic C. albicans through stepwise processes, such as pathogen recognition, cytokine production, and immune cell phagocytosis. Herein, the current advances in the interactions between C. albicans and host mucosal immune defenses have been summarized to improve understanding on the immune mechanisms against fungal infections.
Collapse
Affiliation(s)
- Yujie Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
| | - Yu L. Lei
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Biao Ren
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Investigation of Stress Response Genes in Antimicrobial Resistant Pathogens Sampled from Five Countries. Processes (Basel) 2021. [DOI: 10.3390/pr9060927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Pathogens, which survive from stressed environmental conditions and evolve with antimicrobial resistance, cause millions of human diseases every year in the world. Fortunately, the NCBI Pathogen Detection Isolates Browser (NPDIB) collects the detected stress response genes and antimicrobial resistance genes in pathogen isolates sampled around the world. While several studies have been conducted to identify important antimicrobial resistance genes, little work has been done to analyze the stress response genes in the NPDIB database. In order to address this, this work conducted the first comprehensive statistical analysis of the stress response genes from five countries of the major residential continents, including the US, the UK, China, Australia, and South Africa. Principal component analysis was first conducted to project the stress response genes onto a two-dimensional space, and hierarchical clustering was then implemented to identify the outlier (i.e., important) genes that show high occurrences in the historical data from 2010 to 2020. Stress response genes and AMR genes were finally analyzed together to investigate the co-occurring relationship between these two types of genes. It turned out that seven genes were commonly found in all five countries (i.e., arsR, asr, merC, merP, merR, merT, and qacdelta1). Pathogens E. coli and Shigella, Salmonella enterica, and Klebsiella pneumoniae were the major pathogens carrying the stress response genes. The hierarchical clustering result showed that certain stress response genes and AMR genes were grouped together, including golT~golS and mdsB~mdsC, ymgB and mdtM, and qacEdelta1 and sul1. The occurrence analysis showed that the samples containing three stress response genes and three AMR genes had the highest detection frequency in the historical data. The findings of this work on the important stress response genes, along with their connection with AMR genes, could inform future drug development that targets stress response genes to weaken antimicrobial resistance pathogens.
Collapse
|
20
|
Schuster S, Ewald J, Kaleta C. Modeling the energy metabolism in immune cells. Curr Opin Biotechnol 2021; 68:282-291. [PMID: 33770632 DOI: 10.1016/j.copbio.2021.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/16/2021] [Accepted: 03/01/2021] [Indexed: 02/08/2023]
Abstract
In this review, we summarize and briefly discuss various approaches to modeling the metabolism in human immune cells, with a focus on energy metabolism. These approaches include metabolic reconstruction, elementary modes, and flux balance analysis, which are often subsumed under constraint-based modeling. Further approaches are evolutionary game theory and kinetic modeling. Many immune cells such as macrophages show the Warburg effect, meaning that glycolysis is upregulated upon activation. We outline a minimal model for explaining that effect using optimization. The effect of a confrontation with pathogen cells on immunometabolism is highlighted. Models describing the differences between M1 and M2 macrophages, ROS production in neutrophils, and tryptophan metabolism are discussed. Obstacles and future prospects are outlined.
Collapse
Affiliation(s)
- Stefan Schuster
- Department of Bioinformatics, Matthias Schleiden Institute, Friedrich Schiller University Jena, Ernst-Abbe-Pl. 2, 07743 Jena, Germany.
| | - Jan Ewald
- Department of Bioinformatics, Matthias Schleiden Institute, Friedrich Schiller University Jena, Ernst-Abbe-Pl. 2, 07743 Jena, Germany
| | - Christoph Kaleta
- Medical Systems Biology Group, Institute of Experimental Medicine, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
21
|
Thanh Nguyen H, Zhang R, Inokawa N, Oura T, Chen X, Iwatani S, Niimi K, Niimi M, Holmes AR, Cannon RD, Kajiwara S. Candida albicans Bgl2p, Ecm33p, and Als1p proteins are involved in adhesion to saliva-coated hydroxyapatite. J Oral Microbiol 2021; 13:1879497. [PMID: 33628397 PMCID: PMC7889271 DOI: 10.1080/20002297.2021.1879497] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 02/06/2023] Open
Abstract
Introduction: Candida albicans is an opportunistic pathogen that causes oral candidiasis. A previous study showed that Bgl2p and Ecm33p may mediate the interaction between the yeast and saliva-coated hydroxyapatite (SHA; a model for the tooth surface). This study investigated the roles of these cell wall proteins in the adherence of C. albicans to SHA beads. Methods: C. albicans BGL2 and ECM33 null mutants were generated from wild-type strain SC5314 by using the SAT1-flipper gene disruption method. A novel method based on labelling the yeast with Nile red, was used to investigate the adherence. Results: Adhesion of bgl2Δ and ecm33Δ null mutants to SHA beads was 76.4% and 64.8% of the wild-type strain, respectively. Interestingly, the adhesion of the bgl2Δ, ecm33Δ double mutant (87.7%) was higher than that of both single mutants. qRT-PCR analysis indicated that the ALS1 gene was over-expressed in the bgl2Δ, ecm33Δ strain. The triple null mutant showed a significantly reduced adherence to the beads, (37.6%), compared to the wild-type strain. Conclusion: Bgl2p and Ecm33p contributed to the interaction between C. albicans and SHA beads. Deletion of these genes triggered overexpression of the ALS1 gene in the bgl2Δ/ecm33Δ mutant strain, and deletion of all three genes caused a significant decrease in adhesion.
Collapse
Affiliation(s)
- Hoa Thanh Nguyen
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| | - Rouyu Zhang
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| | - Naoki Inokawa
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| | - Takahiro Oura
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| | - Xinyue Chen
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| | - Shun Iwatani
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| | - Kyoko Niimi
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| | - Masakazu Niimi
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| | - Ann Rachel Holmes
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Richard David Cannon
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Susumu Kajiwara
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| |
Collapse
|
22
|
Valand N, Girija UV. Candida Pathogenicity and Interplay with the Immune System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1313:241-272. [PMID: 34661898 DOI: 10.1007/978-3-030-67452-6_11] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Candida species are opportunistic fungal pathogens that are part of the normal skin and mucosal microflora. Overgrowth of Candida can cause infections such as thrush or life-threatening invasive candidiasis in immunocompromised patients. Though Candida albicans is highly prevalent, several non-albicans species are also isolated from nosocomial infections. Candida sp. are over presented in the gut of people with Crohn's disease and certain types of neurological disorders, with hyphal form and biofilms being the most virulent states. In addition, Candida uses several secreted and cell surface molecules such as pH related antigen 1, High affinity glucose transporter, Phosphoglycerate mutase 1 and lipases to establish pathogenicity. A strong innate immune response is elicited against Candida via dendritic cells, neutrophils and macrophages. All three complement pathways are also activated. Production of proinflammatory cytokines IL-10 and IL-12 signal differentiation of CD4+ cells into Th1 and Th2 cells, whereas IL-6, IL-17 and IL-23 induce Th17 cells. Importance of T-lymphocytes is reflected in depleted T-cell count patients being more prone to Candidiasis. Anti- Candida antibodies also play a role against candidiasis using various mechanisms such as targeting virulent enzymes and exhibiting direct candidacidal activity. However, the significance of antibody response during infection remains controversial. Furthermore, some of the Candida strains have evolved molecular strategies to evade the sophisticated host attack by proteolysis of components of immune system and interfering with immune signalling pathways. Emergence of several non-albicans species that are resistant to current antifungal agents makes treatment more difficult. Therefore, deeper insight into interactions between Candida and the host immune system is required for discovery of novel therapeutic options.
Collapse
Affiliation(s)
- Nisha Valand
- Leicester School of Allied Health and Life sciences, Faculty of Health and Life Sciences, De Montfort University, Leicester, UK
| | - Umakhanth Venkatraman Girija
- Leicester School of Allied Health and Life sciences, Faculty of Health and Life Sciences, De Montfort University, Leicester, UK.
| |
Collapse
|
23
|
Verma G, Kumar B, Sharma AK, Yadav AN. Fungicide as Potential Vaccine: Current Research and Future Challenges. Fungal Biol 2021. [DOI: 10.1007/978-3-030-85603-8_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
24
|
Rottmann BG, Singh PK, Singh S, Revankar SG, Chandrasekar PH, Kumar A. Evaluation of Susceptibility and Innate Immune Response in C57BL/6 and BALB/c Mice During Candida albicans Endophthalmitis. Invest Ophthalmol Vis Sci 2020; 61:31. [PMID: 32940660 PMCID: PMC7500134 DOI: 10.1167/iovs.61.11.31] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022] Open
Abstract
Purpose Candida remains the leading cause of fungal endophthalmitis. However, the pathobiology and innate immune responses in this disease are not well characterized. Here, we developed two murine models of candida endophthalmitis and evaluated their disease susceptibility and differential immune response. Methods Endophthalmitis was induced in C57BL/6 (B6) and BALB/c mice by intravitreal injection of Candida albicans (CA). Disease progression was monitored by slit-lamp examination and clinical scoring, followed by retinal function assessment using electroretinography (ERG). Enucleated eyes were used to estimate fungal burden and retinal tissue damage by hematoxylin and eosin and TUNEL staining. The level of inflammatory mediators were determined by quantitative Polymerase Chain Reaction (qPCR) and enzyme-linked immunosorbent assay, whereas neutrophil infiltration was assessed by flow cytometry and immunostaining. Results Intravitreal injection of CA at 6500 colony-forming units resulted in sustained (non-resolving) ocular inflammation in both B6 and BALB/c mice as evidenced by increased levels of inflammatory cytokines (tumor necrosis factor-α, interleukin-1β, and interleukin-6) and chemokine (CXCL2/MIP-2). In both mouse strains, fungal burden peaked at 24 to 48 hours post-infection (hpi) and decreased by 72 to 96 hpi. CA-infected eyes exhibited increased polymorphonuclear neutrophils (PMN) infiltration and retinal tissue damage. Overall retinal function declined rapidly, with a significant reduction in ERG response at 12 hpi and near-total loss by 24 hpi. Differential analyses revealed increased pathology in BALB/c versus B6 mice. Conclusions C. albicans was able to cause endophthalmitis in mice. Although BALB/c mice were found to be more susceptible to CA endophthalmitis, both BALB/c and B6 models could be used to study fungal endophthalmitis and test therapeutic modalities.
Collapse
Affiliation(s)
- Bruce G. Rottmann
- Department of Ophthalmology, Visual, and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Pawan Kumar Singh
- Department of Ophthalmology, Visual, and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Sneha Singh
- Department of Ophthalmology, Visual, and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Sanjay G. Revankar
- Division of Infectious Disease, Department of Internal Medicine, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Pranatharthi H. Chandrasekar
- Division of Infectious Disease, Department of Internal Medicine, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Ashok Kumar
- Department of Ophthalmology, Visual, and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan, United States
| |
Collapse
|
25
|
Zhu H, Zhu N, Peng L, Zhang B, Yu Q, Li M. The inositol polyphosphate kinase Ipk1 transcriptionally regulates mitochondrial functions in Candida albicans. FEMS Yeast Res 2020; 20:5896454. [DOI: 10.1093/femsyr/foaa050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/21/2020] [Indexed: 12/22/2022] Open
Abstract
ABSTRACT
Inositol polyphosphates (IPs) is an important family of signaling molecules that regulate multiple cellular processes, such as chromatin remodeling, transcription and mRNA export. Inositol polyphosphate kinases, as the critical enzymes for production and transformation of IPs, directly determine the intracellular levels of IPs and therefore are involved in many cellular processes. However, its roles in Candida albicans, the leading fungal pathogen in human beings, remain to be investigated. In this study, we identified the inositol polyphosphate kinase Ipk1 in C. albicans and found that it localizes in the nucleus. Moreover, in the ipk1Δ/Δ mutant, the activity of mitochondrial respiratory chain complexes and the mitochondrial function was severely impaired, which were associated with down-regulation of mitochondrial function-related genes revealed by transcription profiling analysis. The ipk1Δ/Δ mutant also displayed hypersensitivity to a series of environmental stresses, such as antifungal drugs, oxidants, cell wall perturbing agents and macrophage attacks, followed by attenuation of virulence in a mouse systematic infection model. These findings firstly reported the importance of inositol polyphosphate kinase Ipk1 in C. albicans, especially its role in mitochondrial function maintenance and pathogenicity.
Collapse
Affiliation(s)
- Hangqi Zhu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Science, Nankai University, No. 94, Weijin Road, Tianjin 300071, P. R. China
| | - Nali Zhu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Science, Nankai University, No. 94, Weijin Road, Tianjin 300071, P. R. China
| | - Liping Peng
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Science, Nankai University, No. 94, Weijin Road, Tianjin 300071, P. R. China
| | - Bing Zhang
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Science, Nankai University, No. 94, Weijin Road, Tianjin 300071, P. R. China
| | - Qilin Yu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Science, Nankai University, No. 94, Weijin Road, Tianjin 300071, P. R. China
| | - Mingchun Li
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Science, Nankai University, No. 94, Weijin Road, Tianjin 300071, P. R. China
| |
Collapse
|
26
|
Su S, Li X, Yang X, Li Y, Chen X, Sun S, Jia S. Histone acetylation/deacetylation in Candida albicans and their potential as antifungal targets. Future Microbiol 2020; 15:1075-1090. [PMID: 32854542 DOI: 10.2217/fmb-2019-0343] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Recently, the incidence of invasive fungal infections has significantly increased. Candida albicans (C. albicans) is the most common opportunistic fungal pathogen that infects humans. The limited number of available antifungal agents and the emergence of drug resistance pose difficulties to treatment, thus new antifungals are urgently needed. Through their functions in DNA replication, DNA repair and transcription, histone acetyltransferases (HATs) and histone deacetylases (HDACs) perform essential functions relating to growth, virulence, drug resistance and stress responses of C. albicans. Here, we summarize the physiological and pathological functions of HATs/HDACs, potential antifungal targets and underlying antifungal compounds that impact histone acetylation and deacetylation. We anticipate this review will stimulate the identification of new HAT/HDAC-related antifungal targets and antifungal agents.
Collapse
Affiliation(s)
- Shan Su
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, People’s Republic of China
| | - Xiuyun Li
- Department of Pharmacy, Qilu Children’s Hospital, Shandong University, Jinan 250022, China
| | - Xinmei Yang
- Department of Clinical Pharmacy, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, Shandong Province, People’s Republic of China
| | - Yiman Li
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, People’s Republic of China
| | - Xueqi Chen
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, People’s Republic of China
| | - Shujuan Sun
- Department of Clinical Pharmacy, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, Shandong Province, People’s Republic of China
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, Shandong Province, People’s Republic of China
| | - Shuang Jia
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, Shandong Province, People’s Republic of China
| |
Collapse
|
27
|
Halder LD, Jo EAH, Hasan MZ, Ferreira-Gomes M, Krüger T, Westermann M, Palme DI, Rambach G, Beyersdorf N, Speth C, Jacobsen ID, Kniemeyer O, Jungnickel B, Zipfel PF, Skerka C. Immune modulation by complement receptor 3-dependent human monocyte TGF-β1-transporting vesicles. Nat Commun 2020; 11:2331. [PMID: 32393780 PMCID: PMC7214408 DOI: 10.1038/s41467-020-16241-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles have an important function in cellular communication. Here, we show that human and mouse monocytes release TGF-β1-transporting vesicles in response to the pathogenic fungus Candida albicans. Soluble β-glucan from C. albicans binds to complement receptor 3 (CR3, also known as CD11b/CD18) on monocytes and induces the release of TGF-β1-transporting vesicles. CR3-dependence is demonstrated using CR3-deficient (CD11b knockout) monocytes generated by CRISPR-CAS9 genome editing and isolated from CR3-deficient (CD11b knockout) mice. These vesicles reduce the pro-inflammatory response in human M1-macrophages as well as in whole blood. Binding of the vesicle-transported TGF-β1 to the TGF-β receptor inhibits IL1B transcription via the SMAD7 pathway in whole blood and induces TGFB1 transcription in endothelial cells, which is resolved upon TGF-β1 inhibition. Notably, human complement-opsonized apoptotic bodies induce production of similar TGF-β1-transporting vesicles in monocytes, suggesting that the early immune response might be suppressed through this CR3-dependent anti-inflammatory vesicle pathway.
Collapse
Affiliation(s)
- Luke D Halder
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany
| | - Emeraldo A H Jo
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany
| | - Mohammad Z Hasan
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany
| | - Marta Ferreira-Gomes
- Department of Cell Biology, Institute of Biochemistry and Biophysics, Friedrich Schiller University, 07745, Jena, Germany
| | - Thomas Krüger
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany
| | - Martin Westermann
- Electron Microscopy Center, University Hospital Jena, 07743, Jena, Germany
| | - Diana I Palme
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany
| | - Günter Rambach
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, A-6020, Innsbruck, Austria
| | - Niklas Beyersdorf
- Institute for Virology and Immunobiology, University of Würzburg, 97070, Würzburg, Germany
| | - Cornelia Speth
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, A-6020, Innsbruck, Austria
| | - Ilse D Jacobsen
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany.,Friedrich Schiller University, 07743, Jena, Germany
| | - Olaf Kniemeyer
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany
| | - Berit Jungnickel
- Department of Cell Biology, Institute of Biochemistry and Biophysics, Friedrich Schiller University, 07745, Jena, Germany
| | - Peter F Zipfel
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany.,Friedrich Schiller University, 07743, Jena, Germany
| | - Christine Skerka
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany.
| |
Collapse
|
28
|
Ewald J, Sieber P, Garde R, Lang SN, Schuster S, Ibrahim B. Trends in mathematical modeling of host-pathogen interactions. Cell Mol Life Sci 2020; 77:467-480. [PMID: 31776589 PMCID: PMC7010650 DOI: 10.1007/s00018-019-03382-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 11/05/2019] [Accepted: 11/12/2019] [Indexed: 12/18/2022]
Abstract
Pathogenic microorganisms entail enormous problems for humans, livestock, and crop plants. A better understanding of the different infection strategies of the pathogens enables us to derive optimal treatments to mitigate infectious diseases or develop vaccinations preventing the occurrence of infections altogether. In this review, we highlight the current trends in mathematical modeling approaches and related methods used for understanding host-pathogen interactions. Since these interactions can be described on vastly different temporal and spatial scales as well as abstraction levels, a variety of computational and mathematical approaches are presented. Particular emphasis is placed on dynamic optimization, game theory, and spatial modeling, as they are attracting more and more interest in systems biology. Furthermore, these approaches are often combined to illuminate the complexities of the interactions between pathogens and their host. We also discuss the phenomena of molecular mimicry and crypsis as well as the interplay between defense and counter defense. As a conclusion, we provide an overview of method characteristics to assist non-experts in their decision for modeling approaches and interdisciplinary understanding.
Collapse
Affiliation(s)
- Jan Ewald
- Matthias Schleiden Institute, Bioinformatics, Friedrich Schiller University Jena, Ernst-Abbe-Platz 2, 07743, Jena, Germany
| | - Patricia Sieber
- Matthias Schleiden Institute, Bioinformatics, Friedrich Schiller University Jena, Ernst-Abbe-Platz 2, 07743, Jena, Germany
| | - Ravindra Garde
- Matthias Schleiden Institute, Bioinformatics, Friedrich Schiller University Jena, Ernst-Abbe-Platz 2, 07743, Jena, Germany
- Max Planck Institute for Chemical Ecology, Hans-Knöll-Str. 8, 07745, Jena, Germany
| | - Stefan N Lang
- Matthias Schleiden Institute, Bioinformatics, Friedrich Schiller University Jena, Ernst-Abbe-Platz 2, 07743, Jena, Germany
| | - Stefan Schuster
- Matthias Schleiden Institute, Bioinformatics, Friedrich Schiller University Jena, Ernst-Abbe-Platz 2, 07743, Jena, Germany.
| | - Bashar Ibrahim
- Matthias Schleiden Institute, Bioinformatics, Friedrich Schiller University Jena, Ernst-Abbe-Platz 2, 07743, Jena, Germany.
- Centre for Applied Mathematics and Bioinformatics, Gulf University for Science and Technology, 32093, Hawally, Kuwait.
| |
Collapse
|
29
|
Janowska-Sejda EI, Lysenko A, Urban M, Rawlings C, Tsoka S, Hammond-Kosack KE. PHI-Nets: A Network Resource for Ascomycete Fungal Pathogens to Annotate and Identify Putative Virulence Interacting Proteins and siRNA Targets. Front Microbiol 2019; 10:2721. [PMID: 31866958 PMCID: PMC6908471 DOI: 10.3389/fmicb.2019.02721] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 11/08/2019] [Indexed: 12/28/2022] Open
Abstract
Interactions between proteins underlie all aspects of complex biological mechanisms. Therefore, methodologies based on complex network analyses can facilitate identification of promising candidate genes involved in phenotypes of interest and put this information into appropriate contexts. To facilitate discovery and gain additional insights into globally important pathogenic fungi, we have reconstructed computationally inferred interactomes using an interolog and domain-based approach for 15 diverse Ascomycete fungal species, across nine orders, specifically Aspergillus fumigatus, Bipolaris sorokiniana, Blumeria graminis f. sp. hordei, Botrytis cinerea, Colletotrichum gloeosporioides, Colletotrichum graminicola, Fusarium graminearum, Fusarium oxysporum f. sp. lycopersici, Fusarium verticillioides, Leptosphaeria maculans, Magnaporthe oryzae, Saccharomyces cerevisiae, Sclerotinia sclerotiorum, Verticillium dahliae, and Zymoseptoria tritici. Network cartography analysis was associated with functional patterns of annotated genes linked to the disease-causing ability of each pathogen. In addition, for the best annotated organism, namely F. graminearum, the distribution of annotated genes with respect to network structure was profiled using a random walk with restart algorithm, which suggested possible co-location of virulence-related genes in the protein–protein interaction network. In a second ‘use case’ study involving two networks, namely B. cinerea and F. graminearum, previously identified small silencing plant RNAs were mapped to their targets. The F. graminearum phenotypic network analysis implicates eight B. cinerea targets and 35 F. graminearum predicted interacting proteins as prime candidate virulence genes for further testing. All 15 networks have been made accessible for download at www.phi-base.org providing a rich resource for major crop plant pathogens.
Collapse
Affiliation(s)
- Elzbieta I Janowska-Sejda
- Department of Biointeractions and Crop Protection, Rothamsted Research, Harpenden, United Kingdom.,Department of Computational and Analytical Sciences, Rothamsted Research, Harpenden, United Kingdom.,Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, London, United Kingdom
| | - Artem Lysenko
- Department of Computational and Analytical Sciences, Rothamsted Research, Harpenden, United Kingdom
| | - Martin Urban
- Department of Biointeractions and Crop Protection, Rothamsted Research, Harpenden, United Kingdom
| | - Chris Rawlings
- Department of Computational and Analytical Sciences, Rothamsted Research, Harpenden, United Kingdom
| | - Sophia Tsoka
- Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, London, United Kingdom
| | - Kim E Hammond-Kosack
- Department of Biointeractions and Crop Protection, Rothamsted Research, Harpenden, United Kingdom
| |
Collapse
|
30
|
Kalia N, Singh J, Kaur M. Immunopathology of Recurrent Vulvovaginal Infections: New Aspects and Research Directions. Front Immunol 2019; 10:2034. [PMID: 31555269 PMCID: PMC6722227 DOI: 10.3389/fimmu.2019.02034] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 08/12/2019] [Indexed: 12/25/2022] Open
Abstract
Recurrent vulvovaginal infections (RVVI), a devastating group of mucosal infection, are severely affecting women's quality of life. Our understanding of the vaginal defense mechanisms have broadened recently with studies uncovering the inflammatory nature of bacterial vaginosis, inflammatory responses against novel virulence factors, innate Type 17 cells/IL-17 axis, neutrophils mediated killing of pathogens by a novel mechanism, and oxidative stress during vaginal infections. However, the pathogens have fine mechanisms to subvert or manipulate the host immune responses, hijack them and use them for their own advantage. The odds of hijacking increases, due to impaired immune responses, the net magnitude of which is the result of numerous genetic variations, present in multiple host genes, detailed in this review. Thus, by underlining the role of the host immune responses in disease etiology, modern research has clarified a major hypothesis shift in the pathophilosophy of RVVI. This knowledge can further be used to develop efficient immune-based diagnosis and treatment strategies for this enigmatic disease conditions. As for instance, plasma-derived MBL replacement, adoptive T-cell, and antibody-based therapies have been reported to be safe and efficacious in infectious diseases. Therefore, these emerging immune-therapies could possibly be the future therapeutic options for RVVI.
Collapse
Affiliation(s)
- Namarta Kalia
- Department of Molecular Biology and Biochemistry, Guru Nanak Dev University, Amritsar, India
| | - Jatinder Singh
- Department of Molecular Biology and Biochemistry, Guru Nanak Dev University, Amritsar, India
| | - Manpreet Kaur
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, India
| |
Collapse
|
31
|
Shivarathri R, Tscherner M, Zwolanek F, Singh NK, Chauhan N, Kuchler K. The Fungal Histone Acetyl Transferase Gcn5 Controls Virulence of the Human Pathogen Candida albicans through Multiple Pathways. Sci Rep 2019; 9:9445. [PMID: 31263212 PMCID: PMC6603162 DOI: 10.1038/s41598-019-45817-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/14/2019] [Indexed: 12/28/2022] Open
Abstract
Fungal virulence is regulated by a tight interplay of transcriptional control and chromatin remodelling. Despite compelling evidence that lysine acetylation modulates virulence of pathogenic fungi such as Candida albicans, the underlying mechanisms have remained largely unexplored. We report here that Gcn5, a paradigm lysyl-acetyl transferase (KAT) modifying both histone and non-histone targets, controls fungal morphogenesis - a key virulence factor of C. albicans. Our data show that genetic removal of GCN5 abrogates fungal virulence in mice, suggesting strongly diminished fungal fitness in vivo. This may at least in part arise from increased susceptibility to killing by macrophages, as well as by other phagocytes such as neutrophils or monocytes. Loss of GCN5 also causes hypersensitivity to the fungicidal drug caspofungin. Caspofungin hypersusceptibility requires the master regulator Efg1, working in concert with Gcn5. Moreover, Gcn5 regulates multiple independent pathways, including adhesion, cell wall-mediated MAP kinase signaling, hypersensitivity to host-derived oxidative stress, and regulation of the Fks1 glucan synthase, all of which play critical roles in virulence and antifungal susceptibility. Hence, Gcn5 regulates fungal virulence through multiple mechanisms, suggesting that specific inhibition of Gcn5 could offer new therapeutic strategies to combat invasive fungal infections.
Collapse
Affiliation(s)
- Raju Shivarathri
- Medical University of Vienna, Max Perutz Labs Vienna, Campus Vienna Biocenter, A-1030, Vienna, Austria
| | - Michael Tscherner
- Medical University of Vienna, Max Perutz Labs Vienna, Campus Vienna Biocenter, A-1030, Vienna, Austria
| | - Florian Zwolanek
- Medical University of Vienna, Max Perutz Labs Vienna, Campus Vienna Biocenter, A-1030, Vienna, Austria
| | | | - Neeraj Chauhan
- Public Health Research Institute, New Jersey Medical School, Rutgers The State University of New Jersey, Newark, NJ, 07103, USA.
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers The State University of New Jersey, Newark, NJ, 07103, USA.
| | - Karl Kuchler
- Medical University of Vienna, Max Perutz Labs Vienna, Campus Vienna Biocenter, A-1030, Vienna, Austria.
| |
Collapse
|
32
|
A natural histone H2A variant lacking the Bub1 phosphorylation site and regulated depletion of centromeric histone CENP-A foster evolvability in Candida albicans. PLoS Biol 2019; 17:e3000331. [PMID: 31226107 PMCID: PMC6613695 DOI: 10.1371/journal.pbio.3000331] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 07/08/2019] [Accepted: 06/03/2019] [Indexed: 12/24/2022] Open
Abstract
Eukaryotes have evolved elaborate mechanisms to ensure that chromosomes segregate with high fidelity during mitosis and meiosis, and yet specific aneuploidies can be adaptive during environmental stress. Here, we identify a chromatin-based system required for inducible aneuploidy in a human pathogen. Candida albicans utilizes chromosome missegregation to acquire tolerance to antifungal drugs and for nonmeiotic ploidy reduction after mating. We discovered that the ancestor of C. albicans and 2 related pathogens evolved a variant of histone 2A (H2A) that lacks the conserved phosphorylation site for kinetochore-associated Bub1 kinase, a key regulator of chromosome segregation. Using engineered strains, we show that the relative gene dosage of this variant versus canonical H2A controls the fidelity of chromosome segregation and the rate of acquisition of tolerance to antifungal drugs via aneuploidy. Furthermore, whole-genome chromatin precipitation analysis reveals that Centromere Protein A/ Centromeric Histone H3-like Protein (CENP-A/Cse4), a centromeric histone H3 variant that forms the platform of the eukaryotic kinetochore, is depleted from tetraploid-mating products relative to diploid parents and is virtually eliminated from cells exposed to aneuploidy-promoting cues. We conclude that genetically programmed and environmentally induced changes in chromatin can confer the capacity for enhanced evolvability via chromosome missegregation.
Collapse
|
33
|
Lang SN, Germerodt S, Glock C, Skerka C, Zipfel PF, Schuster S. Molecular crypsis by pathogenic fungi using human factor H. A numerical model. PLoS One 2019; 14:e0212187. [PMID: 30779817 PMCID: PMC6380567 DOI: 10.1371/journal.pone.0212187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 01/29/2019] [Indexed: 02/02/2023] Open
Abstract
Molecular mimicry is the formation of specific molecules by microbial pathogens to avoid recognition and attack by the immune system of the host. Several pathogenic Ascomycota and Zygomycota show such a behaviour by utilizing human complement factor H to hide in the blood stream. We call this type of mimicry molecular crypsis. Such a crypsis can reach a point where the immune system can no longer clearly distinguish between self and non-self cells. Thus, a trade-off between attacking disguised pathogens and erroneously attacking host cells has to be made. Based on signalling theory and protein-interaction modelling, we here present a mathematical model of molecular crypsis of pathogenic fungi using the example of Candida albicans. We tackle the question whether perfect crypsis is feasible, which would imply that protection of human cells by complement factors would be useless. The model identifies pathogen abundance relative to host cell abundance as the predominant factor influencing successful or unsuccessful molecular crypsis. If pathogen cells gain a (locally) quantitative advantage over host cells, even autoreactivity may occur. Our new model enables insights into the mechanisms of candidiasis-induced sepsis and complement-associated autoimmune diseases.
Collapse
Affiliation(s)
- Stefan N. Lang
- Dept. of Bioinformatics, Friedrich Schiller University Jena, Jena, Germany
| | | | - Christina Glock
- Dept. of Bioinformatics, Friedrich Schiller University Jena, Jena, Germany
| | - Christine Skerka
- Dept. of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | - Peter F. Zipfel
- Dept. of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
- Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany
| | - Stefan Schuster
- Dept. of Bioinformatics, Friedrich Schiller University Jena, Jena, Germany
- * E-mail:
| |
Collapse
|
34
|
Yeh SJ, Yeh CC, Lan CY, Chen BS. Investigating Common Pathogenic Mechanisms between Homo sapiens and Different Strains of Candida albicans for Drug Design: Systems Biology Approach via Two-Sided NGS Data Identification. Toxins (Basel) 2019; 11:toxins11020119. [PMID: 30769958 PMCID: PMC6409619 DOI: 10.3390/toxins11020119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/08/2019] [Accepted: 02/11/2019] [Indexed: 01/15/2023] Open
Abstract
Candida albicans (C. albicans) is the most prevalent fungal species. Although it is a healthy microbiota, genetic and epigenetic alterations in host and pathogen, and microenvironment changes would lead to thrush, vaginal yeast infection, and even hematogenously disseminated infection. Despite the fact that cytotoxicity is well-characterized, few studies discuss the genome-wide genetic and epigenetic molecular mechanisms between host and C. albicans. The aim of this study is to identify drug targets and design a multiple-molecule drug to prevent the infection from C. albicans. To investigate the common and specific pathogenic mechanisms in human oral epithelial OKF6/TERT-2 cells during the C. albicans infection in different strains, systems modeling and big databases mining were used to construct candidate host–pathogen genetic and epigenetic interspecies network (GEIN). System identification and system order detection are applied on two-sided next generation sequencing (NGS) data to build real host–pathogen cross-talk GEINs. Core host–pathogen cross-talk networks (HPCNs) are extracted by principal network projection (PNP) method. By comparing with core HPCNs in different strains of C. albicans, common pathogenic mechanisms were investigated and several drug targets were suggested as follows: orf19.5034 (YBP1) with the ability of anti-ROS; orf19.939 (NAM7), orf19.2087 (SAS2), orf19.1093 (FLO8) and orf19.1854 (HHF22) with high correlation to the hyphae growth and pathogen protein interaction; orf19.5585 (SAP5), orf19.5542 (SAP6) and orf19.4519 (SUV3) with the cause of biofilm formation. Eventually, five corresponding compounds—Tunicamycin, Terbinafine, Cerulenin, Tetracycline and Tetrandrine—with three known drugs could be considered as a potential multiple-molecule drug for therapeutic treatment of C. albicans.
Collapse
Affiliation(s)
- Shan-Ju Yeh
- Laboratory of Control and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Chun-Chieh Yeh
- Laboratory of Control and Systems Biology, Department of Electrical Engineering, 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.
| | - Bor-Sen Chen
- Laboratory of Control and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan.
- Department of Electrical Engineering, Yuan Ze University, Chungli 32003, Taiwan.
| |
Collapse
|
35
|
|
36
|
Nami S, Mohammadi R, Vakili M, Khezripour K, Mirzaei H, Morovati H. Fungal vaccines, mechanism of actions and immunology: A comprehensive review. Biomed Pharmacother 2018; 109:333-344. [PMID: 30399567 DOI: 10.1016/j.biopha.2018.10.075] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/02/2018] [Accepted: 10/14/2018] [Indexed: 11/28/2022] Open
Abstract
Fungal infections include a wide range of opportunistic and invasive diseases. Two of four major fatal diseases in patients with human immunodeficiency virus (HIV) infection are related to the fungal infections, cryptococcosis, and pneumocystosis. Disseminated candidiasis and different clinical forms of aspergillosis annually impose expensive medical costs to governments and hospitalized patients and ultimately lead to high mortality rates. Therefore, urgent implementations are necessary to prevent the expansion of these diseases. Designing an effective vaccine is one of the most important approaches in this field. So far, numerous efforts have been carried out in developing an effective vaccine against fungal infections. Some of these challenges engaged in different stages of clinical trials but none of them could be approved by the United States Food and Drug Administration (FDA). Here, in addition to have a comprehensive overview on the data from studied vaccine programs, we will discuss the immunology response against fungal infections. Moreover, it will be attempted to clarify the underlying immune mechanisms of vaccines targeting different fungal infections that are crucial for designing an effective vaccination strategy.
Collapse
Affiliation(s)
- Sanam Nami
- Department of Medical Mycology and Parasitology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rasoul Mohammadi
- Department of Medical Parasitology and Mycology, School of Medicine/Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahshid Vakili
- Department of Medical Mycology and Parasitology/Invasive Fungi Research Center (IFRC), School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Kimia Khezripour
- Department of Pharmacotherapy, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamid Morovati
- Department of Medical Mycology and Parasitology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
37
|
Zhang X, Li T, Chen X, Wang S, Liu Z. Nystatin enhances the immune response against Candida albicans and protects the ultrastructure of the vaginal epithelium in a rat model of vulvovaginal candidiasis. BMC Microbiol 2018; 18:166. [PMID: 30359236 PMCID: PMC6202846 DOI: 10.1186/s12866-018-1316-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 10/15/2018] [Indexed: 02/06/2023] Open
Abstract
Background Vulvovaginal candidiasis (VVC) is a common infectious disease of the lower genital tract. Nystatin, a polyene fungicidal antibiotic, is used as a topical antifungal agent for VVC treatment. The aim of the current study was to investigate the possible immunomodulatory effects of nystatin on the vaginal mucosal immune response during Candida albicans infection and examine its role in protection of vaginal epithelial cell (VEC) ultrastructure. Results Following infection with C. albicans, IFN-γ and IL-17 levels in VECs were significantly elevated, while the presence of IgG was markedly decreased as compared to uninfected controls (P < 0.05). No significant differences in IL4 expression were observed. After treatment with nystatin, the level of IFN-γ, IL-17 and IgG was dramatically increased in comparison to the untreated group (P < 0.05). Transmission electron microscopy revealed that C. albicans invades the vaginal epithelium by both induced endocytosis and active penetration. Nystatin treatment protects the ultrastructure of the vaginal epithelium. Compared with the untreated C. albicans-infected group, Flameng scores which measure mitochondrial damage of VECs were markedly decreased (P < 0.001) and the number of adhesive and invasive C. albicans was significantly reduced (P < 0.01) after treatment with nystatin. Conclusions Nystatin plays a protective role in the host defense against C. albicans by up-regulating the IFN-γ-related cellular response, the IL-17 signaling pathway and possibly through enhancing VEC-derived IgG-mediated immunity. Furthermore, nystatin notably improves the ultramorphology of the vaginal mucosa, partially through the protection of mitochondria ultrastructure in VECs and inhibition of adhesion and invasion by C. albicans. Together, these effects enhance the immune response of the vaginal mucosa against C. albicans and protect the ultrastructure of vaginal epithelium in VVC rats.
Collapse
Affiliation(s)
- Xu Zhang
- Laboratory of Electron Microscopy, Ultrastructural Pathology Center, Peking University First Hospital, Beijing, 100034, China
| | - Ting Li
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
| | - Xi Chen
- Department of Gynecology Minimally Invasive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
| | - Suxia Wang
- Laboratory of Electron Microscopy, Ultrastructural Pathology Center, Peking University First Hospital, Beijing, 100034, China
| | - Zhaohui Liu
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China.
| |
Collapse
|
38
|
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
|
39
|
Richardson JP, Moyes DL, Ho J, Naglik JR. Candida innate immunity at the mucosa. Semin Cell Dev Biol 2018; 89:58-70. [PMID: 29501618 DOI: 10.1016/j.semcdb.2018.02.026] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/27/2018] [Indexed: 12/17/2022]
Abstract
The tremendous diversity in microbial species that colonise the mucosal surfaces of the human body is only now beginning to be fully appreciated. Distinguishing between the behaviour of commensal microbes and harmful pathogens that reside at mucosal sites in the body is a complex, and exquisitely fine-tuned process central to mucosal health. The fungal pathobiont Candida albicans is frequently isolated from mucosal surfaces with an asymptomatic carriage rate of approximately 60% in the human population. While normally a benign member of the microbiota, overgrowth of C. albicans often results in localised mucosal infection causing morbidity in otherwise healthy individuals, and invasive infection that often causes death in the absence of effective immune defence. C. albicans triggers numerous innate immune responses at mucosal surfaces, and detection of C. albicans hyphae in particular, stimulates the production of antimicrobial peptides, danger-associated molecular patterns and cytokines that function to reduce fungal burdens during infection. This review will summarise our current understanding of innate immune responses to C. albicans at mucosal surfaces.
Collapse
Affiliation(s)
| | - David L Moyes
- Centre for Host-Microbiome Interactions, Mucosal & Salivary Biology Division, Dental Institute, King's College London, UK.
| | - Jemima Ho
- Mucosal & Salivary Biology Division, Dental Institute, King's College London, UK.
| | - Julian R Naglik
- Mucosal & Salivary Biology Division, Dental Institute, King's College London, UK.
| |
Collapse
|
40
|
Dühring S, Ewald J, Germerodt S, Kaleta C, Dandekar T, Schuster S. Modelling the host-pathogen interactions of macrophages and Candida albicans using Game Theory and dynamic optimization. J R Soc Interface 2018; 14:rsif.2017.0095. [PMID: 28701506 PMCID: PMC5550964 DOI: 10.1098/rsif.2017.0095] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/16/2017] [Indexed: 12/21/2022] Open
Abstract
The release of fungal cells following macrophage phagocytosis, called non-lytic expulsion, is reported for several fungal pathogens. On one hand, non-lytic expulsion may benefit the fungus in escaping the microbicidal environment of the phagosome. On the other hand, the macrophage could profit in terms of avoiding its own lysis and being able to undergo proliferation. To analyse the causes of non-lytic expulsion and the relevance of macrophage proliferation in the macrophage–Candida albicans interaction, we employ Evolutionary Game Theory and dynamic optimization in a sequential manner. We establish a game-theoretical model describing the different strategies of the two players after phagocytosis. Depending on the parameter values, we find four different Nash equilibria and determine the influence of the systems state of the host upon the game. As our Nash equilibria are a direct consequence of the model parameterization, we can depict several biological scenarios. A parameter region, where the host response is robust against the fungal infection, is determined. We further apply dynamic optimization to analyse whether macrophage mitosis is relevant in the host–pathogen interaction of macrophages and C. albicans. For this, we study the population dynamics of the macrophage–C. albicans interactions and the corresponding optimal controls for the macrophages, indicating the best macrophage strategy of switching from proliferation to attacking fungal cells.
Collapse
Affiliation(s)
- Sybille Dühring
- Department of Bioinformatics, Friedrich-Schiller-University Jena, Jena, Germany
| | - Jan Ewald
- Department of Bioinformatics, Friedrich-Schiller-University Jena, Jena, Germany
| | - Sebastian Germerodt
- Department of Bioinformatics, Friedrich-Schiller-University Jena, Jena, Germany
| | - Christoph Kaleta
- Research Group Medical Systems Biology, Institute for Experimental Medicine, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Thomas Dandekar
- Biocenter, Department of Bioinformatics, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Stefan Schuster
- Department of Bioinformatics, Friedrich-Schiller-University Jena, Jena, Germany
| |
Collapse
|
41
|
Huang Y, Cao Y, Li J, Liu Y, Zhong W, Li X, Chen C, Hao P. A survey on cellular RNA editing activity in response to Candida albicans infections. BMC Genomics 2018; 19:43. [PMID: 29363428 PMCID: PMC5780849 DOI: 10.1186/s12864-017-4374-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Adenosine-to-Inosine (A-to-I) RNA editing is catalyzed by the adenosine deaminase acting on RNA (ADAR) family of enzymes, which induces alterations in mRNA sequence. It has been shown that A-to-I RNA editing events are of significance in the cell’s innate immunity and cellular response to viral infections. However, whether RNA editing plays a role in cellular response to microorganism/fungi infection has not been determined. Candida albicans, one of the most prevalent human pathogenic fungi, usually act as a commensal on skin and superficial mucosal, but has been found to cause candidiasis in immunosuppression patients. Previously, we have revealed the up-regulation of A-to-I RNA editing activity in response to different types of influenza virus infections. The current work is designed to study the effect of microorganism/fungi infection on the activity of A-to-I RNA editing in infected hosts. Results We first detected and characterized the A-to-I RNA editing events in oral epithelial cells (OKF6) and primary human umbilical vein endothelial cells (HUVEC), under normal growth condition or with C. albicans infection. Eighty nine thousand six hundred forty eight and 60,872 A-to-I editing sites were detected in normal OKF6 and HUVEC cells, respectively. They were validated against the RNA editing databases, DARNED, RADAR, and REDIportal with 50, 80, and 80% success rates, respectively. While over 95% editing sites were detected in Alu regions, among the rest of the editing sites in non repetitive regions, the majority was located in introns and UTRs. The distributions of A-to-I editing activity and editing depth were analyzed during the course of C. albicans infection. While the normalized editing levels of common editing sites exhibited a significant increase, especially in Alu regions, no significant change in the expression of ADAR1 or ADAR2 was observed. Second, we performed further analysis on data from in vivo mouse study with C. albicans infection. One thousand one hundred thirty three and 955 A-to-I editing sites were identified in mouse tongue and kidney tissues, respectively. The number of A-to-I editing events was much smaller than in human epithelial or endothelial cells, due to the lack of Alu elements in mouse genome. Furthermore, during the course of C. albicans infection we observed stable level of A-to-I editing activity in 131 and 190 common editing sites in the mouse tongue and kidney tissues, and found no significant change in ADAR1 or ADAR2 expression (with the exception of ADAR2 displaying a significant increase at 12 h after infection in mouse kidney tissue before returning to normal). Conclusions This work represents the first comprehensive analysis of A-to-I RNA editome in human epithelial and endothelial cells. C. albicans infection of human epithelial and endothelial cells led to the up-regulation of A-to-I editing activities, through a mechanism different from that of viral infections in human hosts. However, the in vivo mouse model with C. albicans infection did not show significant changes in A-to-I editing activities in tongue and kidney tissues. The different results in the mouse model were likely due to the presence of more complex in vivo environments, e.g. circulation and mixed cell types. Electronic supplementary material The online version of this article (10.1186/s12864-017-4374-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yaowei Huang
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 20031, China
| | - Yingying Cao
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 20031, China
| | - Jiarui Li
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100102, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100102, China
| | - Yuanhua Liu
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 20031, China
| | - Wu Zhong
- National Engineering Research Center For the Emergence Drugs, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Xuan Li
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 20032, China.
| | - Chen Chen
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100102, China. .,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100102, China.
| | - Pei Hao
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 20031, China.
| |
Collapse
|
42
|
Zhang W, Yu X, Kwak M, Xu L, Zhang L, Yu Q, Jin JO. Maturation of dendritic cells by pullulan promotes anti-cancer effect. Oncotarget 2018; 7:44644-44659. [PMID: 27341129 PMCID: PMC5190125 DOI: 10.18632/oncotarget.10183] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 05/17/2016] [Indexed: 12/30/2022] Open
Abstract
Previous studies have demonstrated that pullulan, a polysaccharide purified from Aureobasidium pullulans, has immune-stimulatory effects on T and B cells. Moreover, pullulan has been used as a carrier in the delivery of the antigen (Ag) peptide to lymphoid tissues. However, the in vivo effect of pullulan on dendritic cells (DC) has not been well characterized. In this study, we assessed the effect of pullulan on DC activation and anti-cancer immunity. The results showed that the pullulan treatment up-regulated co-stimulatory molecule expression and enhanced pro-inflammatory cytokine production in bone marrow-derived DCs (BMDC) in vitro and in spleen DCs in vivo. Moreover, the combination of ovalbumin (OVA) and pullulan induced OVA antigen-specific T cell activations in vivo. In tumor-bearing mice, pullulan induced the maturation of DCs in spleen and tumor draining lymph node (drLN), and promoted the OVA-specific T cell activation and migration of the T cells into the tumor. In addition, the combination of OVA and pullulan inhibited B16-OVA tumor growth and liver metastasis. The combination of tyrosinase-related protein 2 (TRP2) peptide and pullulan treatment also suppressed B16 melanoma growth. Thus, the results demonstrated that pullulan enhanced DC maturation and function, and it acted as an adjuvant in promoting Ag-specific immune responses in mice. Thus, pullulan could be a new and useful adjuvant for use in therapeutic cancer vaccines.
Collapse
Affiliation(s)
- Wei Zhang
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoqian Yu
- Periodontal Department, Peking University School and Hospital of Stomatology, Beijing, China
| | - Minseok Kwak
- Department of Chemistry, Pukyong National University, Busan, South Korea
| | - Li Xu
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - LiJun Zhang
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qing Yu
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, MA, USA.,Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - Jun-O Jin
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
43
|
Naglik JR, König A, Hube B, Gaffen SL. Candida albicans-epithelial interactions and induction of mucosal innate immunity. Curr Opin Microbiol 2017; 40:104-112. [PMID: 29156234 PMCID: PMC5733685 DOI: 10.1016/j.mib.2017.10.030] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/27/2017] [Accepted: 10/30/2017] [Indexed: 12/24/2022]
Abstract
Candida albicans is a human fungal pathogen that causes millions of mucosal and life-threatening infections annually. C. albicans initially interacts with epithelial cells, resulting in fungal recognition and the formation of hyphae. Hypha formation is critical for host cell damage and immune activation, which are both driven by the secretion of Candidalysin, a recently discovered peptide toxin. Epithelial activation leads to the production of inflammatory mediators that recruit innate immune cells including neutrophils, macrophages and innate Type 17 cells, which together work with epithelial cells to clear the fungal infection. This review will focus on the recent discoveries that have advanced our understanding of C. albicans-epithelial interactions and the induction of mucosal innate immunity.
Collapse
Affiliation(s)
- Julian R Naglik
- Mucosal and Salivary Biology Division, King's College London Dental Institute, London SE1 1UL, United Kingdom.
| | - Annika König
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knoell-Institute, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knoell-Institute, Jena, Germany; Friedrich Schiller University, Jena, Germany; Center for Sepsis Control and Care, University Hospital, Jena, Germany
| | - Sarah L Gaffen
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
44
|
Șular FL, Dobreanu M. Flow Cytometry Assessment of Bacterial and Yeast Induced Oxidative Burst in Peripheral Blood Phagocytes. ACTA MEDICA MARISIENSIS 2017. [DOI: 10.1515/amma-2017-0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
Objective: The aim of this study was to verify in our laboratory conditions the performance criteria of a commercial kit (PhagoburstTM, Glycotope Biotechnology) as described by the producers. We have also partially altered the use of the available kit by introducing a non-opsonized Candida albicans stimulus, in addition to the opsonized Escherichia coli stimulus provided by the manufacturer. Material and methods: The peripheral blood samples of 6 clinically healthy adults were tested in triplicate according to the manufacturer recommendations. The intraassay imprecision as well as the ranges of neutrophil and monocyte burst activation triggered by various stimuli were assessed. Results: The activation range of granulocytes and monocytes was similar to the one described by the producer in the presence of E. coli (granulocytes: 78.45-99.43% versus 99.6-99.95%, average %CV of 1.53% versus 0.1%, monocytes: 54.63-92.33% versus 81.80-96.67, average %CV 6.92% versus 1.1%). The leukocyte range of activation in the presence of non-opsonized C. albicans was comparable to the one triggered by the fMLP (N-formyl-methionyl-leucyl-phenylalanine) stimulus. Conclusion: The intra-assay precision obtained in our laboratory conditions, as well as the ranges of activated leukocytes, are comparable to the ones described by the producer when using E. coli as a stimulus. The present study shows that introducing an extra fungal stimulus for burst oxidation assessment could provide additional information regarding the non-specific cellular immune response, particularly in patients at risk for candidemia.
Collapse
Affiliation(s)
- Floredana-Laura Șular
- Discipline of Laboratory Medicine, University of Medicine and Pharmacy of Tîrgu Mureș, Tîrgu Mureș , Romania
- Central Laboratory, Emergency Clinical County Hospital of Tîrgu Mureș, Tîrgu Mureș , Romania
| | - Minodora Dobreanu
- Discipline of Laboratory Medicine, University of Medicine and Pharmacy of Tîrgu Mureș, Tîrgu Mureș , Romania
- Central Laboratory, Emergency Clinical County Hospital of Tîrgu Mureș, Tîrgu Mureș , Romania
| |
Collapse
|
45
|
Park SC, Kim YM, Lee JK, Kim NH, Kim EJ, Heo H, Lee MY, Lee JR, Jang MK. Targeting and synergistic action of an antifungal peptide in an antibiotic drug-delivery system. J Control Release 2017; 256:46-55. [DOI: 10.1016/j.jconrel.2017.04.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/29/2017] [Accepted: 04/17/2017] [Indexed: 10/19/2022]
|
46
|
M Weerasekera M, H Sissons C, Wong L, A Anderson S, R Holmes A, D Cannon R. Denaturing gradient gel electrophoresis profiles of bacteria from the saliva of twenty four different individuals form clusters that showed no relationship to the yeasts present. Arch Oral Biol 2017; 82:6-10. [PMID: 28577425 DOI: 10.1016/j.archoralbio.2017.05.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 04/25/2017] [Accepted: 05/20/2017] [Indexed: 10/19/2022]
Abstract
OBJECTIVES The aim was to investigate the relationship between groups of bacteria identified by cluster analysis of the DGGE fingerprints and the amounts and diversity of yeast present. METHODS Bacterial and yeast populations in saliva samples from 24 adults were analysed using denaturing gradient gel electrophoresis (DGGE) of the bacteria present and by yeast culture. RESULTS Eubacterial DGGE banding patterns showed considerable variation between individuals. Seventy one different amplicon bands were detected, the band number per saliva sample ranged from 21 to 39 (mean±SD=29.3±4.9). Cluster and principal component analysis of the bacterial DGGE patterns yielded three major clusters containing 20 of the samples. Seventeen of the 24 (71%) saliva samples were yeast positive with concentrations up to 103cfu/mL. Candida albicans was the predominant species in saliva samples although six other yeast species, including Candida dubliniensis, Candida tropicalis, Candida krusei, Candida guilliermondii, Candida rugosa and Saccharomyces cerevisiae, were identified. The presence, concentration, and species of yeast in samples showed no clear relationship to the bacterial clusters. CONCLUSION Despite indications of in vitro bacteria-yeast interactions, there was a lack of association between the presence, identity and diversity of yeasts and the bacterial DGGE fingerprint clusters in saliva. This suggests significant ecological individual-specificity of these associations in highly complex in vivo oral biofilm systems under normal oral conditions.
Collapse
Affiliation(s)
- Manjula M Weerasekera
- Dental Research Group, School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand; Department of Microbiology, Faculty of Medical Sciences,University of Sri Jayewardenepura, Sri Lanka.
| | - Chris H Sissons
- Dental Research Group, School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand
| | - Lisa Wong
- Dental Research Group, School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand
| | - Sally A Anderson
- Dental Research Group, School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand
| | - Ann R Holmes
- Sir John Walsh Research Institute and Department of Oral Sciences, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Richard D Cannon
- Sir John Walsh Research Institute and Department of Oral Sciences, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| |
Collapse
|
47
|
Role of the Inositol Polyphosphate Multikinase Ipk2 in Regulation of Hyphal Development, Calcium Signaling and Secretion in Candida albicans. Mycopathologia 2017; 182:609-623. [DOI: 10.1007/s11046-017-0138-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 04/12/2017] [Indexed: 10/19/2022]
|
48
|
Abstract
Candida albicans is an important etiological agent of superficial and life-threatening infections in individuals with compromised immune systems. To date, we know of several overlapping genetic networks that govern virulence attributes in this fungal pathogen. Classical use of deletion mutants has led to the discovery of numerous virulence factors over the years, and genome-wide functional analysis has propelled gene discovery at an even faster pace. Indeed, a number of recent studies using large-scale genetic screens followed by genome-wide functional analysis has allowed for the unbiased discovery of many new genes involved in C. albicans biology. Here we share our perspectives on the role of these studies in analyzing fundamental aspects of C. albicans virulence properties.
Collapse
Affiliation(s)
- Thabiso E Motaung
- a Agricultural Research Council - Small Grain Institute , Bethlehem , South Africa
| | - Ruan Ells
- b University of the Free Sate , Bloemfontein , South Africa
| | | | | | - Toi J Tsilo
- a Agricultural Research Council - Small Grain Institute , Bethlehem , South Africa.,c Department of Life and Consumer Sciences , University of South Africa , Pretoria , South Africa
| |
Collapse
|
49
|
Carvalho A, Duarte-Oliveira C, Gonçalves SM, Campos A, Lacerda JF, Cunha C. Fungal Vaccines and Immunotherapeutics: Current Concepts and Future Challenges. CURRENT FUNGAL INFECTION REPORTS 2017. [DOI: 10.1007/s12281-017-0272-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
50
|
Herwig L, Rice AJ, Bedbrook CN, Zhang RK, Lignell A, Cahn JKB, Renata H, Dodani SC, Cho I, Cai L, Gradinaru V, Arnold FH. Directed Evolution of a Bright Near-Infrared Fluorescent Rhodopsin Using a Synthetic Chromophore. Cell Chem Biol 2017; 24:415-425. [PMID: 28262559 DOI: 10.1016/j.chembiol.2017.02.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 11/28/2016] [Accepted: 02/01/2017] [Indexed: 12/16/2022]
Abstract
By engineering a microbial rhodopsin, Archaerhodopsin-3 (Arch), to bind a synthetic chromophore, merocyanine retinal, in place of the natural chromophore all-trans-retinal (ATR), we generated a protein with exceptionally bright and unprecedentedly red-shifted near-infrared (NIR) fluorescence. We show that chromophore substitution generates a fluorescent Arch complex with a 200-nm bathochromic excitation shift relative to ATR-bound wild-type Arch and an emission maximum at 772 nm. Directed evolution of this complex produced variants with pH-sensitive NIR fluorescence and molecular brightness 8.5-fold greater than the brightest ATR-bound Arch variant. The resulting proteins are well suited to bacterial imaging; expression and stability have not been optimized for mammalian cell imaging. By targeting both the protein and its chromophore, we overcome inherent challenges associated with engineering bright NIR fluorescence into Archaerhodopsin. This work demonstrates an efficient strategy for engineering non-natural, tailored properties into microbial opsins, properties relevant for imaging and interrogating biological systems.
Collapse
Affiliation(s)
- Lukas Herwig
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91125, USA
| | - Austin J Rice
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91125, USA
| | - Claire N Bedbrook
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91125, USA; Division of Biology and Biological Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91125, USA
| | - Ruijie K Zhang
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91125, USA
| | - Antti Lignell
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91125, USA
| | - Jackson K B Cahn
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91125, USA
| | - Hans Renata
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91125, USA
| | - Sheel C Dodani
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91125, USA
| | - Inha Cho
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91125, USA
| | - Long Cai
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91125, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91125, USA
| | - Frances H Arnold
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91125, USA; Division of Biology and Biological Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91125, USA.
| |
Collapse
|