201
|
Gaziano R, Sabbatini S, Roselletti E, Perito S, Monari C. Saccharomyces cerevisiae-Based Probiotics as Novel Antimicrobial Agents to Prevent and Treat Vaginal Infections. Front Microbiol 2020; 11:718. [PMID: 32373104 PMCID: PMC7186379 DOI: 10.3389/fmicb.2020.00718] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/27/2020] [Indexed: 12/12/2022] Open
Abstract
Vaginal infections affect 70% of women during their lifetimes and account for millions of annual doctors’ visits. These infections are predominantly represented by vulvovaginal candidiasis (VVC) and bacterial vaginosis (BV). Although standard antimicrobial agents remain the major strategy for the prevention and treatment of vaginal infections, both VVC and BV are difficult to treat due to high rates of resistance and recurrence, high probability of complications, and negative effects on the vaginal microbiota. This review focuses on a new approach of yeast-based probiotics for the prevention and/or treatment of these common vaginal infections.
Collapse
Affiliation(s)
- Roberta Gaziano
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Samuele Sabbatini
- Department of Medicine, Medical Microbiology Section, University of Perugia, Perugia, Italy
| | - Elena Roselletti
- Department of Medicine, Medical Microbiology Section, University of Perugia, Perugia, Italy
| | - Stefano Perito
- Department of Medicine, Medical Microbiology Section, University of Perugia, Perugia, Italy
| | - Claudia Monari
- Department of Medicine, Medical Microbiology Section, University of Perugia, Perugia, Italy
| |
Collapse
|
202
|
The Paralogous Transcription Factors Stp1 and Stp2 of Candida albicans Have Distinct Functions in Nutrient Acquisition and Host Interaction. Infect Immun 2020; 88:IAI.00763-19. [PMID: 32094252 DOI: 10.1128/iai.00763-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 02/14/2020] [Indexed: 01/06/2023] Open
Abstract
Nutrient acquisition is a central challenge for all organisms. For the fungal pathogen Candida albicans, utilization of amino acids has been shown to be critical for survival, immune evasion, and escape, while the importance of catabolism of host-derived proteins and peptides in vivo is less well understood. Stp1 and Stp2 are paralogous transcription factors (TFs) regulated by the Ssy1-Ptr3-Ssy5 (SPS) amino acid sensing system and have been proposed to have distinct, if uncertain, roles in protein and amino acid utilization. We show here that Stp1 is required for proper utilization of peptides but has no effect on amino acid catabolism. In contrast, Stp2 is critical for utilization of both carbon sources. Commensurate with this observation, we found that Stp1 controls a very limited set of genes, while Stp2 has a much more extensive regulon that is partly dependent on the Ssy1 amino acid sensor (amino acid uptake and catabolism) and partly Ssy1 independent (genes associated with filamentous growth, including the regulators UME6 and SFL2). The ssy1Δ/Δ and stp2Δ/Δ mutants showed reduced fitness in a gastrointestinal (GI) colonization model, yet induced greater damage to epithelial cells and macrophages in a manner that was highly dependent on the growth status of the fungal cells. Surprisingly, the stp1Δ/Δ mutant was better able to colonize the gut but the mutation had no effect on host cell damage. Thus, proper protein and amino acid utilization are both required for normal host interaction and are controlled by an interrelated network that includes Stp1 and Stp2.
Collapse
|
203
|
ElBaradei A. A decade after the emergence of Candida auris: what do we know? Eur J Clin Microbiol Infect Dis 2020; 39:1617-1627. [PMID: 32297040 DOI: 10.1007/s10096-020-03886-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 03/30/2020] [Indexed: 12/18/2022]
Abstract
Candida auris is a remarkable emerging pathogen. It has emerged separately, yet simultaneously in different parts of the world, establishing four phylogenetic and geographic distinct clades with a potential fifth clade that was recently reported. C. auris is often perceived as a pathogen in disguise, due to its frequent misidentification and its immune evasion. On the other hand, many of the recovered isolates are multidrug-resistant. In fact, some of these isolates are resistant to the three main antifungal classes: echinocandins, azoles, and polyenes. Moreover, C. auris has the ability to persist and survive on different objects for a long time, aided by different adhering mechanisms including aggregation and biofilm formation, thereby causing outbreaks of invasive infections in hospital settings. However, C. auris ability to maintain its pathogenicity at high temperatures remains among its most unique properties. This is why C. auris represents a challenging threat, and more studies are needed to meet this challenge. This review highlights different characteristics of this emerging yeast with emphasis on its antifungal resistance, its ability to persistent on different surfaces, and its immune evasion capability.
Collapse
Affiliation(s)
- Amira ElBaradei
- Department of Microbiology and Immunology, Faculty of Pharmacy and Drug Manufacturing, Pharos University in Alexandria, Alexandria, Egypt. .,Alexandria University Hospital, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
204
|
The Interactions between the Antimicrobial Peptide P-113 and Living Candida albicans Cells Shed Light on Mechanisms of Antifungal Activity and Resistance. Int J Mol Sci 2020; 21:ijms21072654. [PMID: 32290246 PMCID: PMC7178208 DOI: 10.3390/ijms21072654] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/24/2022] Open
Abstract
In the absence of proper immunity, such as in the case of acquired immune deficiency syndrome (AIDS) patients, Candida albicans, the most common human fungal pathogen, may cause mucosal and even life-threatening systemic infections. P-113 (AKRHHGYKRKFH), an antimicrobial peptide (AMP) derived from the human salivary protein histatin 5, shows good safety and efficacy profiles in gingivitis and human immunodeficiency virus (HIV) patients with oral candidiasis. However, little is known about how P-113 interacts with Candida albicans or its degradation by Candida-secreted proteases that contribute to the fungi’s resistance. Here, we use solution nuclear magnetic resonance (NMR) methods to elucidate the molecular mechanism of interactions between P-113 and living Candida albicans cells. Furthermore, we found that proteolytic cleavage of the C-terminus prevents the entry of P-113 into cells and that increasing the hydrophobicity of the peptide can significantly increase its antifungal activity. These results could help in the design of novel antimicrobial peptides that have enhanced stability in vivo and that can have potential therapeutic applications.
Collapse
|
205
|
Poh SE, Goh JPZ, Fan C, Chua W, Gan SQ, Lim PLK, Sharma B, Leavesley DI, Dawson TL, Li H. Identification of Malassezia furfur Secreted Aspartyl Protease 1 (MfSAP1) and Its Role in Extracellular Matrix Degradation. Front Cell Infect Microbiol 2020; 10:148. [PMID: 32328468 PMCID: PMC7161080 DOI: 10.3389/fcimb.2020.00148] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/20/2020] [Indexed: 12/13/2022] Open
Abstract
Malassezia is the most abundant eukaryotic microbial genus on human skin. Similar to many human-residing fungi, Malassezia has high metabolic potential and secretes a plethora of hydrolytic enzymes that can potentially modify and structure the external skin environment. Here we show that the dominant secreted Malassezia protease isolated from cultured Malassezia furfur is an aspartyl protease that is secreted and active at all phases of culture growth. We observed that this protease, herein named as MfSAP1 (M. furfur secreted aspartyl protease 1) has a broader substrate cleavage profile and higher catalytic efficiency than the previously reported protease homolog in Malassezia globosa. We demonstrate that MfSAP1 is capable of degrading a wide range of human skin associated extracellular matrix (ECM) proteins and ECM isolated directly from keratinocytes and fibroblasts. Using a 3-D wound model with primary keratinocytes grown on human de-epidermized dermis, we show that MfSAP1 protease can potentially interfere with wound re-epithelization in an acute wound model. Taken together, our work demonstrates that Malassezia proteases have host-associated substrates and play important roles in cutaneous wound healing.
Collapse
Affiliation(s)
- Si En Poh
- Molecular Engineering Lab, Institute of Bioengineering and Nanotechnology, Agency for Science Technology and Research, Singapore, Singapore
| | - Joleen P Z Goh
- Skin Research Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
| | - Chen Fan
- Skin Research Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
| | - Wisely Chua
- Molecular Engineering Lab, Institute of Bioengineering and Nanotechnology, Agency for Science Technology and Research, Singapore, Singapore
| | - Shi Qi Gan
- Skin Research Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
| | - Priscilla Lay Keng Lim
- Skin Research Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
| | - Bhavya Sharma
- Skin Research Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
| | - David I Leavesley
- Skin Research Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
| | - Thomas L Dawson
- Skin Research Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore.,School of Pharmacy, Department of Drug Discovery, Medical University of South Carolina, Charleston, SC, United States
| | - Hao Li
- Molecular Engineering Lab, Institute of Bioengineering and Nanotechnology, Agency for Science Technology and Research, Singapore, Singapore
| |
Collapse
|
206
|
Sharma Y, Rastogi SK, Ahmedi S, Manzoor N. Antifungal activity of β -citronellol against two non-albicans Candida species. JOURNAL OF ESSENTIAL OIL RESEARCH 2020. [DOI: 10.1080/10412905.2020.1737588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Yamini Sharma
- Medical Mycology Lab, Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Sumit Kumar Rastogi
- Medical Mycology Lab, Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Saiema Ahmedi
- Medical Mycology Lab, Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Nikhat Manzoor
- Medical Mycology Lab, Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
207
|
Singh DK, Tóth R, Gácser A. Mechanisms of Pathogenic Candida Species to Evade the Host Complement Attack. Front Cell Infect Microbiol 2020; 10:94. [PMID: 32232011 PMCID: PMC7082757 DOI: 10.3389/fcimb.2020.00094] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/24/2020] [Indexed: 11/13/2022] Open
Abstract
Candida species are common colonizers of the human skin, vagina, and the gut. As human commensals, Candida species do not cause any notable damage in healthy individuals; however, in certain conditions they can initiate a wide range of diseases such as chronic disseminated candidiasis, endocarditis, vaginitis, meningitis, and endophthalmitis. The incidence of Candida caused infections has increased worldwide, with mortality rates exceeding 70% in certain patient populations. C. albicans, C. glabrata, C. tropicalis, C. parapsilosis, and C. krusei are responsible for more than 90% of Candida-related infections. Interestingly, the host immune response against these closely related fungi varies. As part of the innate immune system, complement proteins play a crucial role in host defense, protecting the host by lysing pathogens or by increasing their phagocytosis by phagocytes through opsonization. This review summarizes interactions of host complement proteins with pathogenic Candida species, including C. albicans and non-albicans Candida species such as C. parapsilosis. We will also highlight the various ways of complement activation, describe the antifungal effects of complement cascades and explore the mechanisms adopted by members of pathogenic Candida species for evading complement attack.
Collapse
Affiliation(s)
| | - Renáta Tóth
- Department of Microbiology, University of Szeged, Szeged, Hungary
| | - Attila Gácser
- Department of Microbiology, University of Szeged, Szeged, Hungary.,MTA-SZTE Lendület Mycobiome Research Group, University of Szeged, Szeged, Hungary
| |
Collapse
|
208
|
Fidel PL, Yano J, Esher SK, Noverr MC. Applying the Host-Microbe Damage Response Framework to Candida Pathogenesis: Current and Prospective Strategies to Reduce Damage. J Fungi (Basel) 2020; 6:jof6010035. [PMID: 32168864 PMCID: PMC7151217 DOI: 10.3390/jof6010035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 12/16/2022] Open
Abstract
Disease is a complex outcome that can occur as a result of pathogen-mediated damage, host-mediated damage or both. This has led to the revolutionary concept of the damage response framework (DRF) that defines microbial virulence as a function of host immunity. The DRF outlines six scenarios (classes) of host damage or beneficial outcomes, depending on the microbe and the strength of the immune response. Candida albicans is uniquely adapted to its human host and can exist as either a commensal, colonizing various anatomical sites without causing notable damage, or as a pathogen, with the ability to cause a diverse array of diseases, ranging from mucosal to invasive systemic infections that result in varying levels of microbe-mediated and/or host-mediated damage. We recently categorized six different forms of candidiasis (oropharyngeal, hematogenous, intra-abdominal, gastrointestinal, denture stomatitis, and vulvovaginitis) into independent DRF classes, supporting a contemporary view of unique mechanisms of pathogenesis for these Candida infections. In this review, we summarize the evidence for the pathogenesis of these various forms of candidiasis in the context of the DRF with the further intent to provide insights into strategies to achieve a level of host response or outcome otherwise, that limits host damage.
Collapse
Affiliation(s)
- Paul L. Fidel
- Center of Excellence in Oral and Craniofacial Biology, Louisiana State University Health Sciences Center School of Dentistry, New Orleans, LA 70119, USA;
- Correspondence: ; Tel.: +1-504-941-8425
| | - Junko Yano
- Center of Excellence in Oral and Craniofacial Biology, Louisiana State University Health Sciences Center School of Dentistry, New Orleans, LA 70119, USA;
| | - Shannon K. Esher
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA; (S.K.E.); (M.C.N.)
| | - Mairi C. Noverr
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA; (S.K.E.); (M.C.N.)
| |
Collapse
|
209
|
Pellon A, Sadeghi Nasab SD, Moyes DL. New Insights in Candida albicans Innate Immunity at the Mucosa: Toxins, Epithelium, Metabolism, and Beyond. Front Cell Infect Microbiol 2020; 10:81. [PMID: 32195196 PMCID: PMC7062647 DOI: 10.3389/fcimb.2020.00081] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/18/2020] [Indexed: 12/16/2022] Open
Abstract
The mucosal surfaces of the human body are challenged by millions of microbes on a daily basis. Co-evolution with these microbes has led to the development of plastic mechanisms in both host and microorganisms that regulate the balance between preserving beneficial microbes and clearing pathogens. Candida albicans is a fungal pathobiont present in most healthy individuals that, under certain circumstances, can become pathogenic and cause everything from mild mucosal infections to life-threatening systemic diseases. As an essential part of the innate immunity in mucosae, epithelial cells elaborate complex immune responses that discriminate between commensal and pathogenic microbes, including C. albicans. Recently, several significant advances have been made identifying new pieces in the puzzle of host-microbe interactions. This review will summarize these advances in the context of our current knowledge of anti-Candida mucosal immunity, and their impact on epithelial immune responses to this fungal pathogen.
Collapse
Affiliation(s)
- Aize Pellon
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Shervin Dokht Sadeghi Nasab
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - David L Moyes
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| |
Collapse
|
210
|
Abstract
Yeast resistance to antifungal drugs is a major public health issue. Fungal adhesion onto the host mucosal surface is still a partially unknown phenomenon that is modulated by several actors among which fibronectin plays an important role. Targeting the yeast adhesion onto the mucosal surface could lead to potentially highly efficient treatments. In this work, we explored the effect of fibronectin on the nanomotion pattern of different Candida albicans strains by atomic force microscopy (AFM)-based nanomotion detection and correlated the cellular oscillations to the yeast adhesion onto epithelial cells. Preliminary results demonstrate that strongly adhering strains reduce their nanomotion activity upon fibronectin exposure whereas low adhering Candida remain unaffected. These results open novel avenues to explore cellular reactions upon exposure to stimulating agents and possibly to monitor in a rapid and simple manner adhesive properties of C. albicans.
Collapse
|
211
|
Abstract
Candida albicans has remained the main etiological agent of candidiasis, challenges clinicians with high mortality and morbidity. The emergence of resistance to antifungal drugs, toxicity and lower efficacy have all contributed to an urgent need to develop alternative drugs aiming at novel targets in C. albicans. Targeting the production of virulence factors, which are essential processes for infectious agents, represents an attractive substitute for the development of newer anti-infectives. The present review highlights the recent developments made in the understanding of the pathogenicity of C. albicans. Production of hydrolytic enzymes, morphogenesis and biofilm formation, along with their molecular and metabolic regulation in Candida are discussed with regard to the development of novel antipathogenic drugs against candidiasis. Over the last decade, candidiasis has remained a major problematic disease worldwide. In spite of the existence of many antifungal drugs, the treatment of such diseases has still remained unsuccessful due to drug inefficacy. Therefore, there is a need to discover antifungals with different modes of action, such as antipathogenic drugs against Candida albicans. Here, we describe how various types of virulence factors such as proteinase, phospholipase, hemolysin, adhesion, morphogenesis and biofilm formation, could be targeted to develop novel therapeutics. We can inhibit production of these virulence factors by controlling their molecular/metabolic regulation.
Collapse
|
212
|
Yi Y. Functional crosstalk between non-canonical caspase-11 and canonical NLRP3 inflammasomes during infection-mediated inflammation. Immunology 2020; 159:142-155. [PMID: 31630388 PMCID: PMC6954705 DOI: 10.1111/imm.13134] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/25/2019] [Accepted: 10/15/2019] [Indexed: 12/11/2022] Open
Abstract
Inflammation is a part of the body's immune response for protection against pathogenic infections and other cellular damages; however, chronic inflammation is a major cause of various diseases. One key step in the inflammatory response is the activation of inflammasomes, intracellular protein complexes comprising pattern recognition receptors and other inflammatory molecules. The role of the NLRP3 inflammasome in inflammatory responses has been extensively investigated; however, the caspase-11 inflammasome has been recently identified and has been classified as a 'non-canonical' inflammasome, and emerging studies have highlighted its role in inflammatory responses. Because the ligands and the mechanisms for the activation of these two inflammasomes are different, studies to date have separately described their roles, although recent studies have reported the functional cooperation between these two inflammasomes during an inflammatory response. This review discusses the studies investigating the functional crosstalk between non-canonical caspase-11 and canonical NLRP3 inflammasomes in the context of inflammatory responses; moreover, it provides insight for the development of novel anti-inflammatory therapeutics to prevent and treat infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Young‐Su Yi
- Department of Pharmaceutical and Biomedical EngineeringCheongju UniversityCheongjuKorea
| |
Collapse
|
213
|
Shaban S, Patel M, Ahmad A. Improved efficacy of antifungal drugs in combination with monoterpene phenols against Candida auris. Sci Rep 2020; 10:1162. [PMID: 31980703 PMCID: PMC6981193 DOI: 10.1038/s41598-020-58203-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 01/13/2020] [Indexed: 11/09/2022] Open
Abstract
Emergence of Candida auris has been described as a global health threat due to its ability to cause invasive infections with high mortality rate and multidrug resistance. Novel drugs and therapies are required to target this organism and its pathogenicity. Anti-virulence approach and combination therapy have been proposed as alternatives in recent years. This study evaluated the virulence factors in C. auris, combination antifungal activity of phenolic compounds with antifungal drugs and determined effect of the most active compound on positive pathogenicity markers of C. auris. Antifungal susceptibility profile of 25 clinical isolates of C. auris against antifungal agents as well as against phenolic compounds was obtained using CLSI guidelines. Combination of the most active phenolic compound with antifungal drugs was determined. Effect of carvacrol on the virulence factors was also studied. Carvacrol was the most active phenol with median MIC of 125 µg/ml and its combination with fluconazole, amphotericin B, nystatin and caspofungin resulted synergistic and additive effects in 68%, 64%, 96% and 28%, respectively. Combination also reduced the MIC values of the drugs. All test strains showed adherence ability to epithelial cells and 96% of strains produced proteinase. None of the strains produced hyphae and phospholipase. At low concentrations, carvacrol significantly inhibited the adherence ability and proteinase production (both p < 0.01). Carvacrol has antifungal and anti-virulence activity against C. auris. It also showed an enhanced antifungal activity in combination with antifungal agents. Therefore it has potential to be developed into a novel antifungal agent.
Collapse
Affiliation(s)
- Siham Shaban
- Clinical Microbiology and Infectious Diseases, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - Mrudula Patel
- Clinical Microbiology and Infectious Diseases, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa.,Infection Control, Charlotte Maxeke Johannesburg Academic Hospital, National Health Laboratory Service, Johannesburg, 2193, South Africa
| | - Aijaz Ahmad
- Clinical Microbiology and Infectious Diseases, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa. .,Infection Control, Charlotte Maxeke Johannesburg Academic Hospital, National Health Laboratory Service, Johannesburg, 2193, South Africa.
| |
Collapse
|
214
|
Romo JA, Kumamoto CA. On Commensalism of Candida. J Fungi (Basel) 2020; 6:E16. [PMID: 31963458 PMCID: PMC7151168 DOI: 10.3390/jof6010016] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 01/09/2023] Open
Abstract
Candida species are both opportunistic fungal pathogens and common members of the human mycobiome. Over the years, the main focus of the fungal field has been on understanding the pathogenic potential and disease manifestation of these organisms. Therefore, understanding of their commensal lifestyle, interactions with host epithelial barriers, and initial transition into pathogenesis is less developed. In this review, we will describe the current knowledge on the commensal lifestyle of these fungi, how they are able to adhere to and colonize host epithelial surfaces, compete with other members of the microbiota, and interact with the host immune response, as well as their transition into opportunistic pathogens by invading the gastrointestinal epithelium.
Collapse
Affiliation(s)
| | - Carol A. Kumamoto
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA 02111, USA;
| |
Collapse
|
215
|
Vila T, Sultan AS, Montelongo-Jauregui D, Jabra-Rizk MA. Oral Candidiasis: A Disease of Opportunity. J Fungi (Basel) 2020; 6:jof6010015. [PMID: 31963180 PMCID: PMC7151112 DOI: 10.3390/jof6010015] [Citation(s) in RCA: 191] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 12/14/2022] Open
Abstract
Oral candidiasis, commonly referred to as “thrush,” is an opportunistic fungal infection that commonly affects the oral mucosa. The main causative agent, Candida albicans, is a highly versatile commensal organism that is well adapted to its human host; however, changes in the host microenvironment can promote the transition from one of commensalism to pathogen. This transition is heavily reliant on an impressive repertoire of virulence factors, most notably cell surface adhesins, proteolytic enzymes, morphologic switching, and the development of drug resistance. In the oral cavity, the co-adhesion of C. albicans with bacteria is crucial for its persistence, and a wide range of synergistic interactions with various oral species were described to enhance colonization in the host. As a frequent colonizer of the oral mucosa, the host immune response in the oral cavity is oriented toward a more tolerogenic state and, therefore, local innate immune defenses play a central role in maintaining Candida in its commensal state. Specifically, in addition to preventing Candida adherence to epithelial cells, saliva is enriched with anti-candidal peptides, considered to be part of the host innate immunity. The T helper 17 (Th17)-type adaptive immune response is mainly involved in mucosal host defenses, controlling initial growth of Candida and inhibiting subsequent tissue invasion. Animal models, most notably the mouse model of oropharyngeal candidiasis and the rat model of denture stomatitis, are instrumental in our understanding of Candida virulence factors and the factors leading to host susceptibility to infections. Given the continuing rise in development of resistance to the limited number of traditional antifungal agents, novel therapeutic strategies are directed toward identifying bioactive compounds that target pathogenic mechanisms to prevent C. albicans transition from harmless commensal to pathogen.
Collapse
Affiliation(s)
- Taissa Vila
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; (T.V.); (A.S.S.); (D.M.-J.)
| | - Ahmed S. Sultan
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; (T.V.); (A.S.S.); (D.M.-J.)
| | - Daniel Montelongo-Jauregui
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; (T.V.); (A.S.S.); (D.M.-J.)
| | - Mary Ann Jabra-Rizk
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; (T.V.); (A.S.S.); (D.M.-J.)
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
- Correspondence: ; Tel.: +1-410-706-0508; Fax: +1-410-706-0519
| |
Collapse
|
216
|
Hizkiyahu R, Baumfeld Y, Paz Levy D, Lanxner Battat T, Imterat M, Weintraub AY. Antepartum vaginal Candida colonization and the risk for obstetrical tears. J Matern Fetal Neonatal Med 2020; 35:75-79. [PMID: 31937157 DOI: 10.1080/14767058.2020.1712701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Objective: Vaginal Candida colonization is very common during pregnancy. An association between Candida colonization and obstetrical tears has not yet been investigated. We investigated whether vaginal Candida colonization during pregnancy is associated with an increased risk for obstetrical tears.Study design: A prospective cohort study was undertaken between the years 2014-2016, comparing pregnancy and delivery characteristics of women with and without Candida colonization during pregnancy. Clinical characteristics of Candida positive women and those with normal vaginal flora were collected. To test the statistical significance of the categorical variables, the chi-square test or Fisher's exact test were used, where appropriate. For continuous variables, the Student's t-test was used.Results: During the study period, 102 women with vaginal Candida during pregnancy (of whom 70% had Candida albicans species) and 102 controls with normal vaginal flora were included in the analysis. No significant differences were observed between Candida positive women and those with normal vaginal flora. Pregnancy and delivery outcomes were comparable and no increased risk of obstetrical tears was found in the Candida group.Conclusions: Vaginal colonization with Candida species during pregnancy was not found to be associated with an increased risk of obstetrical tears.
Collapse
Affiliation(s)
- Ranit Hizkiyahu
- Department of Obstetrics and Gynecology, Faculty of Health Sciences, Soroka University Medical Center, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Yael Baumfeld
- Department of Obstetrics and Gynecology, Faculty of Health Sciences, Soroka University Medical Center, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Dorit Paz Levy
- Department of Obstetrics and Gynecology, Faculty of Health Sciences, Soroka University Medical Center, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Talya Lanxner Battat
- Department of Obstetrics and Gynecology, Faculty of Health Sciences, Soroka University Medical Center, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Majdi Imterat
- Department of Obstetrics and Gynecology, Faculty of Health Sciences, Soroka University Medical Center, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Adi Y Weintraub
- Department of Obstetrics and Gynecology, Faculty of Health Sciences, Soroka University Medical Center, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| |
Collapse
|
217
|
Serine Protease Inhibitors-New Molecules for Modification of Polymeric Biomaterials. Biomolecules 2020; 10:biom10010082. [PMID: 31947983 PMCID: PMC7023003 DOI: 10.3390/biom10010082] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/25/2019] [Accepted: 12/31/2019] [Indexed: 12/20/2022] Open
Abstract
Three serine protease inhibitors (AEBSF, soy inhibitor, α1-antitrypsin) were covalently immobilized on the surface of three polymer prostheses with the optimized method. The immobilization efficiency ranged from 11 to 51%, depending on the chosen inhibitor and biomaterial. The highest activity for all inhibitors was observed in the case of immobilization on the surface of the polyester Uni-Graft prosthesis, and the preparations obtained showed high stability in the environment with different pH and temperature values. Modification of the Uni-Graft prosthesis surface with the synthetic AEBSF inhibitor and human α1-antitrypsin inhibited the adhesion and multiplication of Staphylococcus aureus subs. aureus ATCC® 25923TM and Candida albicans from the collection of the Department of Genetics and Microbiology, UMCS. Optical profilometry analysis indicated that, after the immobilization process on the surface of AEBSF-modified Uni-Graft prostheses, there were more structures with a high number of protrusions, while the introduction of modifications with a protein inhibitor led to the smoothing of their surface.
Collapse
|
218
|
Genome Assemblies of Two Rare Opportunistic Yeast Pathogens: Diutina rugosa (syn. Candida rugosa) and Trichomonascus ciferrii (syn. Candida ciferrii). G3-GENES GENOMES GENETICS 2019; 9:3921-3927. [PMID: 31575637 PMCID: PMC6893180 DOI: 10.1534/g3.119.400762] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Infections caused by opportunistic yeast pathogens have increased over the last years. These infections can be originated by a large number of diverse yeast species of varying incidence, and with distinct clinically relevant phenotypic traits, such as different susceptibility profiles to antifungal drugs, which challenge diagnosis and treatment. Diutina rugosa (syn. Candida rugosa) and Trichomonascus ciferrii (syn. Candida ciferrii) are two opportunistic rare yeast pathogens, which low incidence (< 1%) limits available clinical experience. Furthermore, these yeasts have elevated Minimum Inhibitory Concentration (MIC) levels to at least one class of antifungal agents. This makes it more difficult to manage their infections, and thus they are associated with high rates of mortality and clinical failure. With the aim of improving our knowledge on these opportunistic pathogens, we assembled and annotated their genomes. A phylogenomics approach revealed that genes specifically duplicated in each of the two species are often involved in transmembrane transport activities. These genomes and the reconstructed complete catalog of gene phylogenies and homology relationships constitute useful resources for future studies on these pathogens.
Collapse
|
219
|
Payne M, Weerasinghe H, Tedja I, Andrianopoulos A. A unique aspartyl protease gene expansion in Talaromyces marneffei plays a role in growth inside host phagocytes. Virulence 2019; 10:277-291. [PMID: 30880596 PMCID: PMC6527018 DOI: 10.1080/21505594.2019.1593776] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/05/2019] [Accepted: 03/05/2019] [Indexed: 11/05/2022] Open
Abstract
Aspartyl proteases are a widely represented class of proteolytic enzymes found in eukaryotes and retroviruses. They have been associated with pathogenicity in a range of disease-causing microorganisms. The dimorphic human-pathogenic fungus Talaromyces marneffei has a large expansion of these proteases identified through genomic analyses. Here we characterize the expansion of these genes (pop - paralogue of pep) and their role in T. marneffei using computational and molecular approaches. Many of the genes in this monophyletic family show copy number variation and positive selection despite the preservation of functional regions and possible redundancy. We show that the expression profile of these genes differs and some are expressed during intracellular growth in the host. Several of these proteins have distinctive localization as well as both additive and epistatic effects on the formation of yeast cells during macrophage infections. The data suggest that this is a recently evolved aspartyl protease gene family which affects intracellular growth and contributes to the pathogenicity of T. marneffei.
Collapse
Affiliation(s)
- Michael Payne
- Genetics, Genomics and Systems Biology, School of BioSciences, University of Melbourne, Australia
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Harshini Weerasinghe
- Genetics, Genomics and Systems Biology, School of BioSciences, University of Melbourne, Australia
| | - Irma Tedja
- Genetics, Genomics and Systems Biology, School of BioSciences, University of Melbourne, Australia
| | - Alex Andrianopoulos
- Genetics, Genomics and Systems Biology, School of BioSciences, University of Melbourne, Australia
| |
Collapse
|
220
|
Ribas CR, Rymovicz AUM, Rosa RT, Peña LC, Bianchini LF, Rosa EAR. Increments in virulence of Candida albicans induced by androgenic anabolic steroids. Steroids 2019; 152:108501. [PMID: 31545962 DOI: 10.1016/j.steroids.2019.108501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 09/16/2019] [Indexed: 11/25/2022]
Affiliation(s)
- Cristina Rauen Ribas
- Brazilian Army. 20(th) Armored Infantry Battalion, Brazil; Xenobiotics Research Unit, School of Life Sciences, The Pontifical Catholic University of Paraná, Brazil
| | | | - Rosimeire Takaki Rosa
- Xenobiotics Research Unit, School of Life Sciences, The Pontifical Catholic University of Paraná, Brazil
| | - Lorena Carolina Peña
- Xenobiotics Research Unit, School of Life Sciences, The Pontifical Catholic University of Paraná, Brazil
| | - Luiz Fernando Bianchini
- Xenobiotics Research Unit, School of Life Sciences, The Pontifical Catholic University of Paraná, Brazil
| | | |
Collapse
|
221
|
Lam S, Zuo T, Ho M, Chan FKL, Chan PKS, Ng SC. Review article: fungal alterations in inflammatory bowel diseases. Aliment Pharmacol Ther 2019; 50:1159-1171. [PMID: 31648369 DOI: 10.1111/apt.15523] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/08/2019] [Accepted: 09/11/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Emerging data suggest that alterations in gut fungi may be associated with the pathogenesis of inflammatory bowel disease (IBD). In healthy individuals, gut commensal fungi act synergistically with other members of the microbiota to maintain homeostasis but their role in IBD is less clear. AIM To review the role of gut fungi and their trans-kingdom interactions with bacteria in IBD METHODS: A literature search was conducted on Ovid and Pubmed to select relevant animal and human studies that have reported fungi and IBD. RESULTS There is an increased total fungal load particularly of Candida and Malassezia species in the faeces and mucosa of Crohn's disease patients, and a lower fungal diversity in the faeces of ulcerative colitis patients. Caspase recruitment domain-containing protein (CARD)-9 polymorphism in Crohn's disease patients favours Malassezia colonisation that worsens gut inflammation. Diet high in carbohydrates increased the total abundance of Candida species, whereas protein-rich diet had the opposite effect. Anti-fungal therapies are mostly used to treat Candida albicans or Histoplasma capsulatum infections in IBD, whereas pilot studies of supplementing fungal probiotics Saccharomycopsis fibuligera, Saccharomyces boulardii and Saccharomyces cerevisiae CNCM I-3856 strain showed therapeutic effects in IBD. CONCLUSIONS Gut fungi are altered in patients with Crohn's disease and ulcerative colitis. Modulation of the fungal microbiota can be considered as a therapeutic approach for IBD. Future research should focus on understanding how the fungal microbiota interacts with other components of the gut microbiota in association with the pathogenesis and development of IBD.
Collapse
Affiliation(s)
- Siu Lam
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Department of Medicine and Therapeutics, Institute of Digestive Disease, LKS Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China.,Department of Microbiology, The Chinese University of Hong Kong, Hong Kong, China
| | - Tao Zuo
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Department of Medicine and Therapeutics, Institute of Digestive Disease, LKS Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Martin Ho
- Department of Life Sciences, Imperial College London, London, UK
| | - Francis K L Chan
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Department of Medicine and Therapeutics, Institute of Digestive Disease, LKS Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Paul K S Chan
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Department of Microbiology, The Chinese University of Hong Kong, Hong Kong, China
| | - Siew C Ng
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Department of Medicine and Therapeutics, Institute of Digestive Disease, LKS Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
222
|
Uyar S, Duman A, Tan A, Akbay Harmandar F, İnal G, Köker G, Şahintürk Y, Bostan F, Çekin AH. Black esophagus. TURKISH JOURNAL OF GASTROENTEROLOGY 2019; 30:986-987. [PMID: 31767554 DOI: 10.5152/tjg.2019.181019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Seyit Uyar
- Departmant of Internal Medicine, Antalya Training and Research Hospital, Antalya, Turkey
| | - Adil Duman
- Departmant of Gastroenterology, Antalya Training and Research Hospital, Antalya, Turkey
| | - Ayşe Tan
- Departmant of Internal Medicine, Antalya Training and Research Hospital, Antalya, Turkey
| | - Ferda Akbay Harmandar
- Departmant of Gastroenterology, Antalya Training and Research Hospital, Antalya, Turkey
| | - Gülşah İnal
- Departmant of Pathology, Antalya Training and Research Hospital, Antalya, Turkey
| | - Gökhan Köker
- Departmant of Internal Medicine, Antalya Training and Research Hospital, Antalya, Turkey
| | - Yasin Şahintürk
- Departmant of Internal Medicine, Antalya Training and Research Hospital, Antalya, Turkey
| | - Feyzi Bostan
- Departmant of Internal Medicine, Antalya Training and Research Hospital, Antalya, Turkey
| | - Ayhan Hilmi Çekin
- Departmant of Gastroenterology, Antalya Training and Research Hospital, Antalya, Turkey
| |
Collapse
|
223
|
Bandara HMHN, Samaranayake LP. Viral, bacterial, and fungal infections of the oral mucosa: Types, incidence, predisposing factors, diagnostic algorithms, and management. Periodontol 2000 2019; 80:148-176. [PMID: 31090135 DOI: 10.1111/prd.12273] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
For millions of years, microbiota residing within us, including those in the oral cavity, coexisted in a harmonious symbiotic fashion that provided a quintessential foundation for human health. It is now clear that disruption of such a healthy relationship leading to microbial dysbiosis causes a wide array of infections, ranging from localized, mild, superficial infections to deep, disseminated life-threatening diseases. With recent advances in research, diagnostics, and improved surveillance we are witnessing an array of emerging and re-emerging oral infections and orofacial manifestations of systemic infections. Orofacial infections may cause significant discomfort to the patients and unnecessary economic burden. Thus, the early recognition of such infections is paramount for holistic patient management, and oral clinicians have a critical role in recognizing, diagnosing, managing, and preventing either new or old orofacial infections. This paper aims to provide an update on current understanding of well-established and emerging viral, bacterial, and fungal infections manifesting in the human oral cavity.
Collapse
Affiliation(s)
| | - Lakshman P Samaranayake
- Department of Oral and Craniofacial Health Sciences, College of Dental Medicine, University of Sharjah, Sharjah, UAE
| |
Collapse
|
224
|
Saito H, Shodo R, Yamazaki K, Katsura K, Ueki Y, Nakano T, Oshikane T, Yamana N, Tanabe S, Utsunomiya S, Ohta A, Abe E, Kaidu M, Sasamoto R, Aoyama H. The association between oral candidiasis and severity of chemoradiotherapy-induced dysphagia in head and neck cancer patients: A retrospective cohort study. Clin Transl Radiat Oncol 2019; 20:13-18. [PMID: 31737796 PMCID: PMC6849117 DOI: 10.1016/j.ctro.2019.10.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 10/21/2019] [Accepted: 10/26/2019] [Indexed: 11/27/2022] Open
Abstract
Oral candidiasis (OC) aggravated dysphagia in chemoradiation for HNC. OC patients required higher doses of opioids. Early diagnosis of chemoradiation-associated OC seems difficult. Antifungal prophylaxis may reduce the severity of mucositis and dysphagia.
Background and purpose Concurrent chemoradiotherapy (CCRT) for head and neck cancer (HNC) is a risk factor for oral candidiasis (OC). As Candida spp. are highly virulent, we conducted a retrospective study to determine whether OC increases the severity of dysphagia related to mucositis in HNC patients. Patients and methods We retrospectively analyzed the cases of consecutive patients with carcinomas of the oral cavity, pharynx, and larynx who underwent CCRT containing cisplatin (CDDP) at our hospital. The diagnosis of OC was based on gross mucosal appearance. We performed a multivariate analysis to determine whether OC was associated with the development of grade 3 dysphagia in the Radiation Therapy Oncology Group (RTOG) Acute Toxicity Criteria. The maximum of the daily opioid doses was compared between the patients with and without OC. Results We identified 138 HNC patients. OC was observed in 51 patients (37%). By the time of their OC diagnosis, 19 (37%) had already developed grade 3 dysphagia. Among the 30 patients receiving antifungal therapy, 12 (40%) showed clinical deterioration. In the multivariate analysis, OC was independently associated with grade 3 dysphagia (OR 2.75; 95%CI 1.22–6.23; p = 0.015). The patients with OC required significantly higher morphine-equivalent doses of opioids (45 vs. 30 mg/day; p = 0.029). Conclusion Candida infection causes refractory dysphagia. It is worth investigating whether antifungal prophylaxis reduces severe dysphagia related to candidiasis.
Collapse
Affiliation(s)
- Hirotake Saito
- Department of Radiation Oncology, Niigata University Medical and Dental Hospital, 1-754 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8520, Japan
| | - Ryusuke Shodo
- Departments of Otolaryngology Head and Neck Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Keisuke Yamazaki
- Department of Otolaryngology Head and Neck Surgery, Niigata University Medical and Dental Hospital, 1-754 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8520, Japan
| | - Kouji Katsura
- Department of Oral Radiology, Niigata University Medical and Dental Hospital, 1-754 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8520, Japan
| | - Yushi Ueki
- Departments of Otolaryngology Head and Neck Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Toshimichi Nakano
- Department of Radiology and Radiation Oncology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Tomoya Oshikane
- Department of Radiology and Radiation Oncology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Nobuko Yamana
- Department of Radiation Oncology, Niigata University Medical and Dental Hospital, 1-754 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8520, Japan
| | - Satoshi Tanabe
- Department of Radiation Oncology, Niigata University Medical and Dental Hospital, 1-754 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8520, Japan
| | - Satoru Utsunomiya
- Department of Radiologic Technology, Niigata University Graduate School of Health Sciences, 2-746 Asahimachi-dori, Chuo-ku, Niigata 951-8518, Japan
| | - Atsushi Ohta
- Department of Radiology and Radiation Oncology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Eisuke Abe
- Department of Radiology and Radiation Oncology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Motoki Kaidu
- Department of Radiology and Radiation Oncology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Ryuta Sasamoto
- Department of Radiologic Technology, Niigata University Graduate School of Health Sciences, 2-746 Asahimachi-dori, Chuo-ku, Niigata 951-8518, Japan
| | - Hidefumi Aoyama
- Department of Radiology and Radiation Oncology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| |
Collapse
|
225
|
Roselletti E, Monari C, Sabbatini S, Perito S, Vecchiarelli A, Sobel JD, Cassone A. A Role for Yeast/Pseudohyphal Cells of Candida albicans in the Correlated Expression of NLRP3 Inflammasome Inducers in Women With Acute Vulvovaginal Candidiasis. Front Microbiol 2019; 10:2669. [PMID: 31803172 PMCID: PMC6873873 DOI: 10.3389/fmicb.2019.02669] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 11/01/2019] [Indexed: 12/29/2022] Open
Abstract
In acute vulvovaginal candidiasis (VVC), the fungus Candida albicans activates inflammasome receptors of vaginal epithelial cells through the production of virulence and immuno-inflammatory factors. Here, we show that in VVC patients, genes encoding some of the above factors (SAP2, SAP5, SAP6, ECE1, and HWP1) are expressed in a correlated fashion. Cytological observations pointed out that pseudohyphal filaments with yeast cells are dominant at the acidic vaginal pH, and this is coupled with co-expression, at roughly similar level, of SAP2, a typical yeast and ECE1, a typical hyphae-associated genes. In contrast, vigorous hyphal growth dominated at the neutral vaginal pH of mice experimentally infected with C. albicans isolates from VVC subjects, and this is coupled with a high ratio of ECE1 to SAP2 expression. We suggest that the pseudohyphal rather than true hyphal cells of C. albicans play a critical role in VVC, possibly through the activity of multiple inflammasome inducers.
Collapse
Affiliation(s)
- Elena Roselletti
- Medical Microbiology Section, Department of Medicine, University of Perugia, Perugia, Italy
| | - Claudia Monari
- Medical Microbiology Section, Department of Medicine, University of Perugia, Perugia, Italy
| | - Samuele Sabbatini
- Medical Microbiology Section, Department of Medicine, University of Perugia, Perugia, Italy
| | - Stefano Perito
- Medical Microbiology Section, Department of Medicine, University of Perugia, Perugia, Italy
| | - Anna Vecchiarelli
- Medical Microbiology Section, Department of Medicine, University of Perugia, Perugia, Italy
| | - Jack D Sobel
- School of Medicine, Wayne State University, Detroit, MI, United States
| | - Antonio Cassone
- Polo d'Innovazione di Genomica, Genetica e Biologia, University of Siena, Siena, Italy
| |
Collapse
|
226
|
Wang SH, Chen CC, Lee CH, Chen XA, Chang TY, Cheng YC, Young JJ, Lu JJ. Fungicidal and anti-biofilm activities of trimethylchitosan-stabilized silver nanoparticles against Candida species in zebrafish embryos. Int J Biol Macromol 2019; 143:724-731. [PMID: 31734360 DOI: 10.1016/j.ijbiomac.2019.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 09/16/2019] [Accepted: 10/01/2019] [Indexed: 02/06/2023]
Abstract
Herein, positively surface-charged silver nanoparticles (AgNPs) capped with trimethylchitosan nitrate (TMCN) were synthesized using an environmentally friendly method. Nano-sized TMCN-AgNPs (~80 nm) with high zeta potential (>30 mV) provide sufficient static repulsion to stabilize colloid AgNPs in aqueous solutions without aggregation for >3 months. In in vitro cell cycle assays, TMCN-AgNPs showed low cytotoxicity towards L929 cells. A microdilution inhibition assay demonstrated the antifungal potential of TMCN-AgNPs, with a minimum inhibitory concentration of 0.06 mM against Candida tropicalis ATCC 750, and 0.46 mM against both Candida albicans ATCC 76615 and Candida glabrata ATCC 15545. Moreover, the addition of TMCN-AgNPs at 0.23 mM significantly reduced biofilm formation in 96-well plates with C. albicans and C. tropicalis. Importantly, when zebrafish eggs were infected with Candida cells, 0.23 mM TMCN-AgNPs greatly diminished the amount of biofilm on eggs and rescued the survival of embryos by up to 70%.
Collapse
Affiliation(s)
- Shao-Hung Wang
- Department of Microbiology, Immunology and Biopharmaceuticals, National Chiayi University, Chiayi City, Taiwan
| | - Cheng-Cheung Chen
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City, Taiwan
| | - Chih-Hua Lee
- Department of Laboratory Medicine, Chang-Gung Memorial Hospital Linkou, Taoyuan City, Taiwan
| | - Xin-An Chen
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City, Taiwan
| | - Tein-Yao Chang
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City, Taiwan
| | - Yi-Chuan Cheng
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Jenn-Jong Young
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City, Taiwan.
| | - Jang-Jih Lu
- Department of Laboratory Medicine, Chang-Gung Memorial Hospital Linkou, Taoyuan City, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan; Department of Medicine, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.
| |
Collapse
|
227
|
Silymarin, a Popular Dietary Supplement Shows Anti- Candida Activity. Antibiotics (Basel) 2019; 8:antibiotics8040206. [PMID: 31683548 PMCID: PMC6963672 DOI: 10.3390/antibiotics8040206] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/17/2019] [Accepted: 10/30/2019] [Indexed: 01/13/2023] Open
Abstract
Silymarin is a complex of plant-derived compounds obtained from the seed shells of the milk thistle (Silybum marianum). It is used in medicine primarily to protect the liver. The mixture contains mainly flavonolignans, with silybin as a paramount bioactive component of the extract. This article presents the potential health benefits for silymarin as an antifungal drug against five references strains: C. albicans, C. glabrata, C. parapsilosis, C. tropicalis, and C. krusei with MIC (minimum inhibitory concentration) values ranging from 30 to 300 µg/mL. Additionally, this study revealed that the compound suppressed the growth of cells of most of the tested clinical Candida albicans strains with MIC values between 30 and 1200 µg/mL. Based on the fractional inhibitory concentration index (FICI), the combination of silymarin with antifungal drugs caspofungin, fluconazole, and amphotericin B did not significantly change the MIC values for the tested Candida strains. Furthermore, no antagonistic reactions were observed in any combination of drugs. In addition, this substance shows anti-virulence properties including the destabilization of mature biofilm and the inhibition of the secretion of hydrolases. qRT-PCR-based experiments demonstrated that the SAP4 gene involved in virulence was downregulated by silymarin. These results indicate completely new advantages of dietary supplementation with this natural plant extract.
Collapse
|
228
|
Li X, Wu X, Gao Y, Hao L. Synergistic Effects and Mechanisms of Combined Treatment With Harmine Hydrochloride and Azoles for Resistant Candida albicans. Front Microbiol 2019; 10:2295. [PMID: 31749766 PMCID: PMC6843067 DOI: 10.3389/fmicb.2019.02295] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 09/20/2019] [Indexed: 01/23/2023] Open
Abstract
Several studies have demonstrated the significant antiviral, antimicrobial, antiplasmodial, antioxidative, antifungal, antimutagenic, and antitumor properties of harmine hydrochloride (HMH). The main objective of the present study was to investigate the antifungal effects and underlying mechanisms of HMH when combined with azoles to determine whether such combinations act in a synergistic manner. As a result, we found that HMH exhibits synergistic antifungal effects in combination with azoles against resistant Candida albicans (C. albicans) planktonic cells, as well as resistant C. albicans biofilm in the early stage. Antifungal potential of HMH with fluconazole was also explored in vivo using an invertebrate model. Our results suggest that HMH combined with azoles is synergistic against resistant C. albicans in vitro and in vivo. No synergy is seen with azole sensitive C. albicans strains nor with other Candida species. Such synergistic mechanisms on resistance C. albicans are involved in increasing intracellular azoles, inhibiting hyphal growth, disturbing cytosolic calcium concentration, and inducing apoptosis of resistant C. albicans cells.
Collapse
Affiliation(s)
- Xiuyun Li
- Department of Pharmacy, Qilu Children's Hospital, Shandong University, Jinan, China
| | - Xuexin Wu
- Department of Pharmacy, Qilu Children's Hospital, Shandong University, Jinan, China
| | - Yan Gao
- Department of Pharmacy, Qilu Children's Hospital, Shandong University, Jinan, China
| | - Lina Hao
- Department of Pharmacy, Qilu Children's Hospital, Shandong University, Jinan, China
| |
Collapse
|
229
|
Shrief R, Zaki MES, El-Sehsah EM, Ghaleb S, Mofreh M. Study of Antifungal Susceptibility, Virulence Genes and Biofilm Formation in Candida albicans. Open Microbiol J 2019. [DOI: 10.2174/1874285801913010241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background:
Candida albicans has emerged as an important nosocomial pathogen. The morbidity and mortality associated with this pathogen are related to the presence of virulence genes and antifungal resistance. The objective of the present study was to investigate the prevalence of antifungal resistance, biofilm formation and some virulence genes such as ALS1, PLB1, INT1, SAP1 and HWP1, among clinical isolates of Candida albicans recovered from immunocompromised patients.
Methods:
The study included one hundred C. albicans isolates identified phenotypically and by a molecular technique using Polymerase Chain Reaction (PCR). The identified C. albicans was further subjected to antifungal study by the microdilution method, biofilm study and molecular study for virulence genes by PCR.
Results:
The resistance to antifungal drugs, fluconazole, caspofungin and itraconazole was 8% for each of them and for amphotericin B, it was 9%. The prevalence of the studied virulence genes was HWP1 77%, INT1 72%, ALS1 65%, SAP1 65% and PLB1 52%. The biofilm capacity was identified by the microplate method in 58% of C. albicans. The OD was intense in 20 isolates, moderate in 21 isolates and mild in 17 isolates. There was a statistically significant increase in the prevalence of the studied virulence genes INT1, ALS1, HWP1, SAP1 and PLB1 among biofilm forming C. albicans as compared to non-biofilm forming isolates (P=0.0001). Additionally, the resistance to fluconazole, itraconazole and caspofungin was statistically, significantly higher in C. albicans with the capacity to form biofilm as compared to non-biofilm forming C. albicans.
Conclusion:
The present study highlights the prevalence of resistance to antifungal drugs among C. albicans which are not uncommon. Moreover, there was a high prevalence of INT1, ALS1, HWP1, SAP1 and PLB1 genes in C. albicans. The resistance to antifungal drugs was common among isolates with the capacity to form the biofilm. There was an association between the biofilm formation and virulence genes.
Collapse
|
230
|
Evaluation of the Antifungal and Wound-Healing Properties of a Novel Peptide-Based Bioadhesive Hydrogel Formulation. Antimicrob Agents Chemother 2019; 63:AAC.00888-19. [PMID: 31332066 DOI: 10.1128/aac.00888-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/14/2019] [Indexed: 12/25/2022] Open
Abstract
Oral candidiasis (OC) caused by the fungal pathogen Candida albicans is the most common opportunistic infection in immunocompromised populations. The dramatic increase in resistance to common antifungal agents has emphasized the importance of identifying alternative therapeutic options. Antimicrobial peptides have emerged as promising drug candidates due to their antimicrobial properties; specifically, histatin-5 (Hst-5), a peptide naturally produced and secreted by human salivary glands, has demonstrated potent activity against C. albicans However, as we previously demonstrated vulnerability for Hst-5 to proteolysis by C. albicans proteolytic enzymes at specific amino acid residues, a new variant (K11R-K17R) was designed with amino acid substitutions at the identified cleavage sites. The new resistant peptide demonstrated no cytotoxicity to erythrocytes or human oral keratinocytes. To evaluate the potential of the new peptide for clinical application, we utilized our FDA-approved polymer-based bioadhesive hydrogel as a delivery system and developed a therapeutic formulation specifically designed for oral topical application. The new formulation was demonstrated to be effective against C. albicans strains resistant to the traditional antifungals, and the in vitro therapeutic efficacy was found to be comparable to that of the common topical antifungal agents in clinical use. Importantly, in addition to its antifungal properties, our findings also demonstrated that the new peptide variant induces cell proliferation and rapid cell migration of human oral keratinocytes, indicative of wound healing properties. The findings from this study support the progression of the novel formulation as a therapeutic agent against oral candidiasis, as well as a therapeutic modality for promoting wound healing.
Collapse
|
231
|
Kart D, Yabanoglu Ciftci S, Nemutlu E. Altered metabolomic profile of dual-species biofilm: Interactions between Proteus mirabilis and Candida albicans. Microbiol Res 2019; 230:126346. [PMID: 31563763 DOI: 10.1016/j.micres.2019.126346] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/18/2019] [Accepted: 09/21/2019] [Indexed: 01/04/2023]
Abstract
In this study, we aimed to determine the interspecies interactions between Proteus mirabilis and Candida albicans. Mono and dual-species biofilms were grown in a microtiter plate and metabolomic analysis of the biofilms was performed. The effects of togetherness of two species on the expression levels of candidal virulence genes and urease and swarming activities of P.mirabilis were investigated. The growth of C.albicans was inhibited by P.mirabilis whereas the growth and swarming activity of P.mirabilis were increased by C.albicans. The inhibition of Candida cell growth was found to be biofilm specific. The alteration was not detected in urease activity. The expressions of EFG1, HWP1 and SAP2 genes were significantly down-regulated, however, LIP1 was upregulated by P.mirabilis. In the presence of P.mirabilis carbonhydrates, amino acids, polyamine and lipid metabolisms were altered in C.albicans. Interestingly, the putrescine level was increased up to 230 fold in dual-species biofilm compared to monospecies C.albicans biofilm. To our knowledge, this is the first study to investigate the impact of each microbial pathogen on the dual microbial environment by integration of metabolomic data.
Collapse
Affiliation(s)
- Didem Kart
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Microbiology, Sıhhiye, Ankara, Turkey.
| | - Samiye Yabanoglu Ciftci
- Hacettepe University, Faculty of Pharmacy, Department of Biochemistry, Sıhhiye, Ankara, Turkey
| | - Emirhan Nemutlu
- Hacettepe University, Faculty of Pharmacy, Department of Analytical Chemistry, Sıhhiye, Ankara, Turkey
| |
Collapse
|
232
|
Coad BR, Michl TD, Bader CA, Baranger J, Giles C, Gonçalves GC, Nath P, Lamont-Friedrich SJ, Johnsson M, Griesser HJ, Plush SE. Visualizing Biomaterial Degradation by Candida albicans Using Embedded Luminescent Molecules To Report on Substrate Digestion and Cellular Uptake of Hydrolysate. ACS APPLIED BIO MATERIALS 2019; 2:3934-3941. [DOI: 10.1021/acsabm.9b00520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Bryan R. Coad
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia
- School of Agriculture, Food & Wine, University of Adelaide, Adelaide 5000, Australia
| | - Thomas D. Michl
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Christie A. Bader
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Joris Baranger
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Carla Giles
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia
- Centre for Aquatic Animal Health & Vaccines, Tasmania Department of Primary Industries Parks Water & Environment, 165 Westbury Road, Prospect, Tasmania 7250, Australia
| | - Giovanna Cufaro Gonçalves
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Pratiti Nath
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | | | - Malin Johnsson
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Hans J. Griesser
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Sally E. Plush
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| |
Collapse
|
233
|
Global Transcriptomic Analysis of the Candida albicans Response to Treatment with a Novel Inhibitor of Filamentation. mSphere 2019; 4:4/5/e00620-19. [PMID: 31511371 PMCID: PMC6739497 DOI: 10.1128/msphere.00620-19] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
These results from whole-genome transcriptional profiling provide further insights into the biological activity and mode of action of a small-molecule inhibitor of C. albicans filamentation. This information will assist in the development of novel antivirulence strategies against C. albicans infections. The opportunistic pathogenic fungus Candida albicans can cause devastating infections in immunocompromised patients. Its ability to undergo a morphogenetic transition from yeast to filamentous forms allows it to penetrate tissues and damage tissues, and the expression of genes associated with a number of pathogenetic mechanisms is also coordinately regulated with the yeast-to-hypha conversion. Therefore, it is widely considered that filamentation represents one of the main virulence factors of C. albicans. We have previously identified N-[3-(allyloxy)-phenyl]-4-methoxybenzamide (compound 9029936) as the lead compound in a series of small-molecule inhibitors of C. albicans filamentation and characterized its activity both in vitro and in vivo. This compound appears to be a promising candidate for the development of alternative antivirulence strategies for the treatment of C. albicans infections. In this study, we performed RNA sequencing analysis of samples obtained from C. albicans cells grown under filament-inducing conditions in the presence or absence of this compound. Overall, treatment with compound 9029936 resulted in 618 upregulated and 702 downregulated genes. Not surprisingly, some of the most downregulated genes included well-characterized genes associated with filamentation and virulence such as SAP5, ECE1 (candidalysin), and ALS3, as well as genes that impact metal chelation and utilization. Gene ontology analysis revealed an overrepresentation of cell adhesion, iron transport, filamentation, biofilm formation, and pathogenesis processes among the genes downregulated during treatment with this leading compound. Interestingly, the top upregulated genes suggested an enhancement of vesicular transport pathways, particularly those involving SNARE interactions. IMPORTANCE These results from whole-genome transcriptional profiling provide further insights into the biological activity and mode of action of a small-molecule inhibitor of C. albicans filamentation. This information will assist in the development of novel antivirulence strategies against C. albicans infections.
Collapse
|
234
|
Adhikari S, Leissa JA, Karlsson AJ. Beyond function: Engineering improved peptides for therapeutic applications. AIChE J 2019. [DOI: 10.1002/aic.16776] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Sayanee Adhikari
- Department of Chemical and Biomolecular Engineering University of Maryland College Park Maryland
| | - Jesse A. Leissa
- Department of Chemical and Biomolecular Engineering University of Maryland College Park Maryland
| | - Amy J. Karlsson
- Department of Chemical and Biomolecular Engineering University of Maryland College Park Maryland
- Fischell Department of Bioengineering University of Maryland College Park Maryland
| |
Collapse
|
235
|
Rizwan K, Khan SA, Ahmad I, Rasool N, Ibrahim M, Zubair M, Jaafar HZ, Manea R. A Comprehensive Review on Chemical and Pharmacological Potential of Viola betonicifolia: A Plant with Multiple Benefits. Molecules 2019; 24:molecules24173138. [PMID: 31470508 PMCID: PMC6749243 DOI: 10.3390/molecules24173138] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/08/2019] [Accepted: 03/16/2019] [Indexed: 11/21/2022] Open
Abstract
Viola betonicifolia (Violaceae) is commonly recognized as “Banafsha” and widely distributed throughout the globe. This plant is of great interest because of its traditional, pharmacological uses. This review mainly emphases on morphology, nutritional composition, and several therapeutic uses, along with pharmacological properties of different parts of this multipurpose plant. Different vegetative parts of this plant (roots, leaves, petioles, and flowers) contained a good profile of essential micro- and macronutrients and are rich source of fat, protein, carbohydrates, and vitamin C. The plant is well known for its pharmacological properties, e.g., antioxidant, antihelminthic, antidepressant, anti-inflammatory, analgesic, and has been reported in the treatment of various neurological diseases. This plant is of high economic value. The plant has potential role in cosmetic industry. This review suggests that V. betonicifolia is a promising source of pharmaceutical agents. This plant is also of significance as ornamental plant, however further studies needed to explore its phytoconstituents and their pharmacological potential. Furthermore, clinical studies are needed to use this plant for benefits of human beings.
Collapse
Affiliation(s)
- Komal Rizwan
- Department of Chemistry, Government College University, Faisalabad 38000, Pakistan.
- Department of Chemistry, Government College Women University, Faisalabad 38000, Pakistan.
| | - Shakeel Ahmad Khan
- Center of Super-Diamond and Advanced Films (COSDAF) and Department of Chemistry, City University of Hong Kong, Hong Kong, China
| | - Ikram Ahmad
- Department of Applied Chemistry, Government College University, Faisalabad 38000, Pakistan
| | - Nasir Rasool
- Department of Chemistry, Government College University, Faisalabad 38000, Pakistan
| | - Muhammad Ibrahim
- Department of Environmental Sciences and Engineering, Government College University, Faisalabad 38000, Pakistan
| | - Muhammad Zubair
- Department of Chemistry, Government College University, Faisalabad 38000, Pakistan
| | - Hawa Ze Jaafar
- Department of Crop Science, Faculty of Agriculture, University Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Rosana Manea
- Faculty of Medicine, Transilvania University of Brasov, 500036 Brasov, Romania
| |
Collapse
|
236
|
Ajdidi A, Sheehan G, Abu Elteen K, Kavanagh K. Assessment of the in vitro and in vivo activity of atorvastatin against Candida albicans. J Med Microbiol 2019; 68:1497-1506. [PMID: 31460860 DOI: 10.1099/jmm.0.001065] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Aim. The aim of this work was to characterize the response of Candida albicans to atorvastatin, and to assess its in vivo antifungal capability.Methodology. The effect of atorvastatin on the growth and viability of C. albicans was assessed. The ability of the statin to alter cell permeability was quantified by measuring amino acid and protein leakage. The response of C. albicans to atorvastatin was assessed using label-free quantitative proteomics. The in vivo antifungal activity of atorvastatin was assessed using Galleria mellonella larvae infected with C. albicans.Results. Atorvastatin inhibited the growth of C. albicans. The atorvastatin-treated cells showed lower ergosterol levels than the controls, demonstrated increased calcofluor staining and released elevated quantities of amino acids and protein. Larvae infected with C. albicans showed a survival rate of 18.1±4.2 % at 144 h. In contrast, larvae administered atorvastatin (9.09 mg kg-1) displayed a survival rate of 60.2±6.4 % (P<0.05). Label-free quantitative proteomics identified 1575 proteins with 2 or more peptides and 465 proteins were differentially abundant (P<0.05). There was an increase in the abundance of enzymes with oxidoreductase and hydrolase activity in atorvastatin-treated cells, and squalene monooxygenase (4.52-fold increase) and lanosterol synthase (2.84-fold increase) were increased in abundance. Proteins such as small heat shock protein 21 (-6.33-fold) and glutathione peroxidase (-2.05-fold) were reduced in abundance.Conclusion. The results presented here indicate that atorvastatin inhibits the growth of C. albicans and is capable of increasing the survival of G. mellonella larvae infected with C. albicans.
Collapse
Affiliation(s)
- Ahmad Ajdidi
- SSPC Research Centre, Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Gerard Sheehan
- SSPC Research Centre, Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Khaled Abu Elteen
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Kevin Kavanagh
- SSPC Research Centre, Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland
| |
Collapse
|
237
|
Pekmezovic M, Mogavero S, Naglik JR, Hube B. Host-Pathogen Interactions during Female Genital Tract Infections. Trends Microbiol 2019; 27:982-996. [PMID: 31451347 DOI: 10.1016/j.tim.2019.07.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/25/2019] [Accepted: 07/22/2019] [Indexed: 12/11/2022]
Abstract
Dysbiosis in the female genital tract (FGT) is characterized by the overgrowth of pathogenic bacterial, fungal, or protozoan members of the microbiota, leading to symptomatic or asymptomatic infections. In this review, we discuss recent advances in studies dealing with molecular mechanisms of pathogenicity factors of Gardnerella vaginalis, Mycoplasma genitalium, Mycoplasma hominis, Neisseria gonorrhoeae, Streptococcus agalactiae, Chlamydia trachomatis, Trichomonas vaginalis, and Candida spp., as well as their interactions with the host and microbiota in the various niches of the FGT. Taking a holistic approach to identifying fundamental commonalities and differences during these infections could help us to better understand reproductive tract health and improve current prevention and treatment strategies.
Collapse
Affiliation(s)
- Marina Pekmezovic
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany
| | - Selene Mogavero
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dental, Oral, and Craniofacial Sciences, King's College London, SE1 1UL, UK
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany; Institute of Microbiology, Friedrich Schiller University, Jena, Germany. @leibniz-hki.de
| |
Collapse
|
238
|
Abstract
Aspartyl proteases are present in various organisms and, among virulent species, are considered major virulence factors. Host tissue and cell damage, hijacking of immune responses, and hiding from innate immune cells are the most common behaviors of fungal secreted proteases enabling pathogen survival and invasion. C. parapsilosis, an opportunistic human-pathogenic fungus mainly threatening low-birth weight neonates and children, possesses three SAPP protein-encoding genes that could contribute to the invasiveness of the species. Our results suggest that SAPP1 and SAPP2, but not SAPP3, influence host evasion by regulating cell damage, phagocytosis, phagosome-lysosome maturation, killing, and cytokine secretion. Furthermore, SAPP1 and SAPP2 also effectively contribute to complement evasion. Candida parapsilosis is an emerging non-albicans Candida species that largely affects low-birth-weight infants and immunocompromised patients. Fungal pathogenesis is promoted by the dynamic expression of diverse virulence factors, with secreted proteolytic enzymes being linked to the establishment and progression of disease. Although secreted aspartyl proteases (Sap) are critical for Candida albicans pathogenicity, their role in C. parapsilosis is poorly elucidated. In the present study, we aimed to examine the contribution of C. parapsilosisSAPP genes SAPP1, SAPP2, and SAPP3 to the virulence of the species. Our results indicate that SAPP1 and SAPP2, but not SAPP3, influence adhesion, host cell damage, phagosome-lysosome maturation, phagocytosis, killing capacity, and cytokine secretion by human peripheral blood-derived macrophages. Purified Sapp1p and Sapp2p were also shown to efficiently cleave host complement component 3b (C3b) and C4b proteins and complement regulator factor H. Additionally, Sapp2p was able to cleave factor H-related protein 5 (FHR-5). Altogether, these data demonstrate the diverse, significant contributions that SAPP1 and SAPP2 make to the establishment and progression of disease by C. parapsilosis through enabling the attachment of the yeast cells to mammalian cells and modulating macrophage biology and disruption of the complement cascade. IMPORTANCE Aspartyl proteases are present in various organisms and, among virulent species, are considered major virulence factors. Host tissue and cell damage, hijacking of immune responses, and hiding from innate immune cells are the most common behaviors of fungal secreted proteases enabling pathogen survival and invasion. C. parapsilosis, an opportunistic human-pathogenic fungus mainly threatening low-birth weight neonates and children, possesses three SAPP protein-encoding genes that could contribute to the invasiveness of the species. Our results suggest that SAPP1 and SAPP2, but not SAPP3, influence host evasion by regulating cell damage, phagocytosis, phagosome-lysosome maturation, killing, and cytokine secretion. Furthermore, SAPP1 and SAPP2 also effectively contribute to complement evasion.
Collapse
|
239
|
Role of Amino Acid Metabolism in the Virulence of Human Pathogenic Fungi. CURRENT CLINICAL MICROBIOLOGY REPORTS 2019. [DOI: 10.1007/s40588-019-00124-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
240
|
Roselletti E, Sabbatini S, Ballet N, Perito S, Pericolini E, Blasi E, Mosci P, Cayzeele Decherf A, Monari C, Vecchiarelli A. Saccharomyces cerevisiae CNCM I-3856 as a New Therapeutic Agent Against Oropharyngeal Candidiasis. Front Microbiol 2019; 10:1469. [PMID: 31354640 PMCID: PMC6637852 DOI: 10.3389/fmicb.2019.01469] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/12/2019] [Indexed: 02/02/2023] Open
Abstract
Oropharyngeal candidiasis is a common opportunistic mucosal infection of the oral cavity, mainly caused by an overgrowth of Candida albicans. This infection can inhibit nutritional intakes and strongly affect quality of life. To date, standard therapeutic strategies involving the administration of antifungal drugs can bring several side effects, not least the emergence of drug-resistant strains. The purpose of this study is to investigate the effectiveness of Saccharomyces cerevisiae CNCM I-3856 (live or inactivated cells) against oropharyngeal candidiasis. Our results show that administration of S. cerevisiae CNCM I-3856 (live or inactivated cells) in the oral cavity of C57BL/6J mice resulted in a protective effect against oropharyngeal candidiasis. The strongest effect was obtained with live S. cerevisiae CNCM I-3856. This was related to: (1) a decrease in C. albicans load in the oral cavity, esophagus, stomach, and duodenum; (2) an early resolution of inflammatory process in the tongue; (3) a marked reduction in C. albicans virulence factors; and (4) a consistent increase in neutrophil antimicrobial capacity. These findings suggest that S. cerevisiae products are potentially beneficial in the treatment of oropharyngeal candidiasis.
Collapse
Affiliation(s)
- Elena Roselletti
- Medical Microbiology Section, Department of Medicine, University of Perugia, Perugia, Italy
| | - Samuele Sabbatini
- Medical Microbiology Section, Department of Medicine, University of Perugia, Perugia, Italy
| | - Nathalie Ballet
- Lesaffre International, Lesaffre Group, Marcq-en-Baroeul, France
| | - Stefano Perito
- Medical Microbiology Section, Department of Medicine, University of Perugia, Perugia, Italy
| | - Eva Pericolini
- Department of Surgical, Medical, Dental and Morphological Sciences With Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisabetta Blasi
- Department of Surgical, Medical, Dental and Morphological Sciences With Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Paolo Mosci
- Internal Medicine Section, Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | | | - Claudia Monari
- Medical Microbiology Section, Department of Medicine, University of Perugia, Perugia, Italy
| | - Anna Vecchiarelli
- Medical Microbiology Section, Department of Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
241
|
Han Z, Kautto L, Meyer W, Chen SCA, Nevalainen H. Effect of peptidases secreted by the opportunistic pathogen Scedosporium aurantiacum on human epithelial cells. Can J Microbiol 2019; 65:814-822. [PMID: 31265796 DOI: 10.1139/cjm-2019-0212] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Peptidases secreted by a clinical high-virulence Scedosporium aurantiacum isolate (strain WM 06.482; CBS 136046) under normoxic and hypoxic conditions were separated via size-exclusion chromatography, and peptidase activities present in each fraction were determined using class-specific substrates. The fractions demonstrating peptidase activity were assessed for their effects on the attachment and viability of A549 human lung epithelial cells in vitro. Of the peptidases detected in the size-exclusion chromatography fractions, the elastase-like peptidase reduced cell viability, the chymotrypsin-like peptidase was associated with cell detachment, and the cysteine peptidases were able to abolish both cell attachment and viability. The loss of cell viability and attachment became more prominent with an increase in the peptidase activity and could also be specifically prevented by addition of class-specific peptidase inhibitors. Our findings indicate that peptidases secreted by S. aurantiacum can breach the human alveolar epithelial cell barrier and, thus, may have a role in the pathobiology of the organism.
Collapse
Affiliation(s)
- Zhiping Han
- Department of Molecular Sciences, Macquarie University, Sydney, Australia
| | - Liisa Kautto
- Department of Molecular Sciences, Macquarie University, Sydney, Australia.,Biomolecular Discovery and Design Research Centre, Macquarie University, Sydney, Australia
| | - Wieland Meyer
- Molecular Mycology Research Laboratory, Centre for Infectious Diseases and Microbiology, Marie Bashir Institute for Infectious Diseases and Biosecurity, Sydney Medical School - Westmead Hospital, The University of Sydney, Westmead Institute for Medical Research, Sydney, Australia
| | - Sharon C-A Chen
- Molecular Mycology Research Laboratory, Centre for Infectious Diseases and Microbiology, Marie Bashir Institute for Infectious Diseases and Biosecurity, Sydney Medical School - Westmead Hospital, The University of Sydney, Westmead Institute for Medical Research, Sydney, Australia.,Centre for Infectious Diseases and Microbiology Laboratory Services, ICPMR, New South Wales Health Pathology, Westmead Hospital, Westmead, NSW, Australia
| | - Helena Nevalainen
- Department of Molecular Sciences, Macquarie University, Sydney, Australia.,Biomolecular Discovery and Design Research Centre, Macquarie University, Sydney, Australia
| |
Collapse
|
242
|
Genotypic Patterns of Secreted Aspartyl Proteinase Gene in Various Candida Species Isolated from Antenatal Women with Vulvovaginal Candidiasis. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2019. [DOI: 10.22207/jpam.13.2.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
243
|
de Jong AW, Hagen F. Attack, Defend and Persist: How the Fungal Pathogen Candida auris was Able to Emerge Globally in Healthcare Environments. Mycopathologia 2019; 184:353-365. [PMID: 31209693 DOI: 10.1007/s11046-019-00351-w] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 05/28/2019] [Indexed: 12/25/2022]
Abstract
Within a decade after its first description, the multidrug-resistant yeast Candida auris has emerged globally as a nosocomial pathogen causing difficult to control outbreaks. This, together with the alarmingly high mortality rate of up to 66% associated with C. auris candidemia, calls for a better understanding of its virulence traits and routes of transmission. Unlike other clinically relevant Candida species, C. auris seems to have the unique ability to be easily transmitted between patients. Although initially thought to express fewer virulence traits than Candida albicans, recent genomic insights suggest C. auris to possess these traits to a much more similar extent. This review highlights the virulence traits C. auris expresses to attack the host, defend itself against antimicrobial agents and to persist within the healthcare environment.
Collapse
Affiliation(s)
- Auke W de Jong
- Department of Medical Mycology, Westerdijk Fungal Biodiversity Institute, Utrecht, The Netherlands
| | - Ferry Hagen
- Department of Medical Mycology, Westerdijk Fungal Biodiversity Institute, Utrecht, The Netherlands.
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands.
- Laboratory of Medical Mycology, Jining No. 1 People's Hospital, Jining, Shandong, People's Republic of China.
| |
Collapse
|
244
|
Basmaciyan L, Bon F, Paradis T, Lapaquette P, Dalle F. " Candida Albicans Interactions With The Host: Crossing The Intestinal Epithelial Barrier". Tissue Barriers 2019; 7:1612661. [PMID: 31189436 PMCID: PMC6619947 DOI: 10.1080/21688370.2019.1612661] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/24/2019] [Accepted: 04/24/2019] [Indexed: 02/08/2023] Open
Abstract
Formerly a commensal organism of the mucosal surfaces of most healthy individuals, Candida albicans is an opportunistic pathogen that causes infections ranging from superficial to the more life-threatening disseminated infections, especially in the ever-growing population of vulnerable patients in the hospital setting. In these situations, the fungus takes advantage of its host following a disturbance in the host defense system and/or the mucosal microbiota. Overwhelming evidence suggests that the gastrointestinal tract is the main source of disseminated C. albicans infections. Major risk factors for disseminated candidiasis include damage to the mucosal intestinal barrier, immune dysfunction, and dysbiosis of the resident microbiota. A better understanding of C. albicans' interaction with the intestinal epithelial barrier will be useful for designing future therapies to avoid systemic candidiasis. In this review, we provide an overview of the current knowledge regarding the mechanisms of pathogenicity that allow the fungus to reach and translocate the gut barrier.
Collapse
Affiliation(s)
- Louise Basmaciyan
- Laboratoire de Parasitologie-Mycologie, Plateforme de Biologie Hospitalo-Universitaire Gérard Mack, Dijon France
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Fabienne Bon
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Tracy Paradis
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Pierre Lapaquette
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Frédéric Dalle
- Laboratoire de Parasitologie-Mycologie, Plateforme de Biologie Hospitalo-Universitaire Gérard Mack, Dijon France
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| |
Collapse
|
245
|
Nichols RG, Peters JM, Patterson AD. Interplay Between the Host, the Human Microbiome, and Drug Metabolism. Hum Genomics 2019; 13:27. [PMID: 31186074 PMCID: PMC6558703 DOI: 10.1186/s40246-019-0211-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 05/21/2019] [Indexed: 02/07/2023] Open
Abstract
The human microbiome is composed of four major areas including intestinal, skin, vaginal, and oral microbiomes, with each area containing unique species and unique functionalities. The human microbiome may be modulated with prebiotics, probiotics, and postbiotics to potentially aid in the treatment of diseases like irritable bowel syndrome, bacterial vaginosis, atopic dermatitis, gingivitis, obesity, or cancer. There is also potential for many of the inhabitants of the human microbiome to directly modulate host gene expression and modulate host detoxifying enzyme activity like cytochrome P450s (CYPs), dehydrogenases, and carboxylesterases. Therefore, the microbiome may be important to consider during drug discovery, risk assessment, and dosing regimens for various diseases given that the human microbiome has been shown to impact host detoxification processes.
Collapse
Affiliation(s)
- Robert G. Nichols
- Department of Veterinary and Biomedical Science, The Pennsylvania State University, University Park, PA 16802 USA
| | - Jeffrey M. Peters
- Department of Veterinary and Biomedical Science, The Pennsylvania State University, University Park, PA 16802 USA
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Science, The Pennsylvania State University, University Park, PA 16802 USA
| |
Collapse
|
246
|
Alburquenque C, Amaro J, Fuentes M, Falconer MA, Moreno C, Covarrubias C, Pinto C, Rodas PI, Bucarey SA, Hermosilla G, Magne F, Tapia CV. Protective effect of inactivated blastoconidia in keratinocytes and human reconstituted epithelium against C. albicans infection. Med Mycol 2019; 57:457-467. [PMID: 30169683 DOI: 10.1093/mmy/myy068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 07/09/2018] [Accepted: 08/23/2018] [Indexed: 12/13/2022] Open
Abstract
Candida albicans is commensal yeast that colonizes skin and mucosa; however, it can become an opportunist pathogen by changing from blastoconidia (commensal form) into hypha (pathogenic form). Each form activates a different cytokines response in epithelial cells. Little is known about the commensal role of C. albicans in the innate immunity. This work studied whether stimulation with C. albicans blastoconidia induces protection in keratinocytes and/or in a reconstituted human epithelium (RHE) infected with C. albicans. For this, inactivated C. albicans blastoconidia was used to stimulate keratinocytes and RHE prior to infection with C. albicans. Blastoconidia induced different cytokine expression profiles; in the case of RHE it decreased interleukin (IL)-1β and IL-10 and increased IL-8, tumor necrosis factor α (TNF-α), and interferon γ (IFN-γ). A significant increase in the expression of human β-defensins (HBD) 2 and HBD3 was observed in blastoconidia stimulated keratinocytes and RHE, associated with impaired growth and viability of C. albicans. Additionally, blastoconidia stimulation decreased the expression of virulence factors in C. albicans that are associated with filamentation (EFG1, CPH1 and NRG1), adhesion (ALS5), and invasion (SAP2). Blastoconidia stimulated RHE was significantly less damaged by C. albicans invasion. These results show that the commensal form of C. albicans would exert a protective effect against self-infection.
Collapse
Affiliation(s)
- Claudio Alburquenque
- Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina Universidad de Chile. Santiago de Chile.,Escuela de Tecnología Médica, Facultad de Ciencias, Universidad Mayor, Santiago de Chile
| | - José Amaro
- Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina Universidad de Chile. Santiago de Chile
| | - Marisol Fuentes
- Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina Universidad de Chile. Santiago de Chile
| | - Mary A Falconer
- Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina Universidad de Chile. Santiago de Chile
| | - Claudia Moreno
- Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina Universidad de Chile. Santiago de Chile
| | - Cristian Covarrubias
- Facultad de Odontología Universidad de Chile. Laboratorio de Nanomateriales. Santiago, Chile
| | - Cristian Pinto
- Unidad de Anatomía Patológica Clínica Dávila, Santiago, Chile
| | - Paula I Rodas
- Laboratorio de Microbiología Médica y Patogénesis Bacteriana, Facultad de Medicina, Universidad Andrés Bello, Concepción, Chile
| | - Sergio A Bucarey
- Centro Biotecnológico Biovetec, Departamento de Ciencias Biológicas Animales. Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago de Chile
| | - Germán Hermosilla
- Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina Universidad de Chile. Santiago de Chile
| | - Fabien Magne
- Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina Universidad de Chile. Santiago de Chile
| | - Cecilia V Tapia
- Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina Universidad de Chile. Santiago de Chile.,Laboratorio Clínica Dávila, Santiago, Chile
| |
Collapse
|
247
|
Wu H, Castanheira P, Faro C, Tang J. Cardosin A endocytosis mediated by integrin leads to lysosome leakage and apoptosis of epithelial cells. Proteins 2019; 87:502-511. [PMID: 30785216 PMCID: PMC11026154 DOI: 10.1002/prot.25672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/28/2019] [Accepted: 02/17/2019] [Indexed: 11/07/2022]
Abstract
Cardosin A is an aspartic protease present in large amount in the pistils of cardoon flowers. This protease is known to contain an -Arg-Gly-Asp- (RGD) motif located on the molecular surface. In this study, we found that isolated recombinant cardosin A attached to human epithelial cells A549, mediated by the binding of its RGD motif to cell surface integrins. The cell bound cardosin A was internalized to endosomes and lysosomes and triggered the permeability of lysosomal membrane leading to apoptosis of the epithelial cells. These events are identical to those observed for three RGD-containing aspartic proteases, Saps 4-6, secreted by Candida albicans. Such a process, which has been called the Trojan Horse mechanism, is believed to benefit the invasion of C. albican into the epithelium of the host. The location of the RGD motifs of cardosin A and Saps 4-6 are on the opposite ends of the homologous three-dimensional structures, suggesting that the Trojan Horse mechanism is insensitive to the RGD position. Current finding also suggests that cardosin A may have a defensive function against the ingestion of cardoon flowers by human, insects, and other herbivores.
Collapse
Affiliation(s)
- Hao Wu
- Protein Studies Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Pedro Castanheira
- The Biocant and Neuroscience and Cell Biology (CNC) department, Coimbra, Portugal and Biocant, Biotechnology Innovation Center, Cantanhede, Portugal
| | - Carlos Faro
- The Biocant and Neuroscience and Cell Biology (CNC) department, Coimbra, Portugal and Biocant, Biotechnology Innovation Center, Cantanhede, Portugal
- The Molecular Biotechnology Unit, Coimbra, Portugal and Biocant, Biotechnology Innovation Center, Cantanhede, Portugal
| | - Jordan Tang
- Protein Studies Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| |
Collapse
|
248
|
Abstract
Background: Adhesion, biofilm formation, yeast-hyphal transition, secretion of enzymes, and hemolytic activity are all considered important factors in Candida tropicalis infection. However, DNA sequence data for this pathogen are limited. In this study, the polymorphism and heterogeneity of genes agglutinin-like sequences (ALS)2, Lipase (LIP)1, LIP4, and secretory aspartyl proteinase tropicalis (SAPT)1-4 as well as the relationship between phenotype and genotype were analyzed. Methods: This study started in August 2013, and ended in July 2017. The complete length of ALS2, LIP1, LIP4, and SAPT1-4 of 68 clinical C. tropicalis isolates was sequenced. Single nucleotide polymorphisms (SNPs) as well as insertions and deletions (indels) were identified within these genes. In addition, phenotypic characteristics of the virulent factors, including adhesion and the secretion of aspartyl proteinases and phospholipases, were determined. Results: There were 73, 24, 17, 16, 13, and 180 SNPs in the genes LIP1, LIP4, SAPT1, SAPT2, SAPT3, and SAPT4, respectively. Furthermore, 209 SNPs were identified in total for the gene ALS2. Interestingly, large fragment deletions and insertions were also found in ALS2. Isolate FXCT 01 obtained from blood had deletions on all 4 sites and showed the lowest adhesion ability on the polymethylpentene surface. In addition, isolates with deletions in the regions 1697 to 1925 and 2073 to 2272 bp displayed relatively low abilities for adhesion and biofilm formation, and this phenotype correlated with the deletions found in ALS2. LIP1, SAPT4, and ALS2 displayed great heterogeneity among the isolates. Large deletions found in gene ALS2 appeared to be associated with the low ability of adhesion and biofilm formation of C. tropicalis. Conclusion: This study might be useful for deeper explorations of gene function and studying the virulent mechanisms of C. tropicalis.
Collapse
|
249
|
Giordani C, Simonetti G, Natsagdorj D, Choijamts G, Ghirga F, Calcaterra A, Quaglio D, De Angelis G, Toniolo C, Pasqua G. Antifungal activity of Mongolian medicinal plant extracts. Nat Prod Res 2019; 34:449-455. [PMID: 31135192 DOI: 10.1080/14786419.2019.1610960] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The in vitro antifungal activity of extracts obtained from 14 medicinal plants of the mongolian flora were investigated by measuring their minimal inhibitory concentration (MIC) against fungi cause of cutaneous diseases such as Candida species, dermatophytes and Malassezia furfur. Among the species examined, Stellaria dichotoma L., Scutellaria scordifolia L. Aquilegia sibirica Fisch. Et Schrenk. and Hyoscyamus niger L. extracts demonstrated antifungal activity against all studied fungi. In particular, S. scordifolia L. methanol extract, obtained at room temperature, showed the best activity against Candida spp., Malassezia furfur and dermatophytes with GMMIC50 values of 22 µg/mL, 64 µg/mL and 32 µg/mL, respectively. The flavones, luteolin and apigenin, identified in S. scordifolia extracts, and rutin identified in S. dichotoma and Hyoscyamus niger L. extracts, could be responsible of the observed antifungal activity.
Collapse
Affiliation(s)
- Cristiano Giordani
- Grupo Productos Naturales Marinos, Facultad de Ciencias Farmacéuticas y Alimentarias, Instituto de Fisica, Universidad de Antioquia, Medellin, Colombia
| | - Giovanna Simonetti
- Department of Public Health and Infectious Diseases, Sapienza Università di Roma, Rome, Italy
| | | | - Gotov Choijamts
- Otoch Manramba University of Mongolia, Ulaanbaatar, Mongolia
| | - Francesca Ghirga
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Andrea Calcaterra
- Department of Chemistry and Technology of Drugs "Department of Excellence 2018-2022", Sapienza Università di Roma, Rome, Italy
| | - Deborah Quaglio
- Department of Chemistry and Technology of Drugs "Department of Excellence 2018-2022", Sapienza Università di Roma, Rome, Italy
| | - Giulia De Angelis
- Department of Environmental Biology, Sapienza Università di Roma, Rome, Italy
| | - Chiara Toniolo
- Department of Environmental Biology, Sapienza Università di Roma, Rome, Italy
| | - Gabriella Pasqua
- Department of Environmental Biology, Sapienza Università di Roma, Rome, Italy
| |
Collapse
|
250
|
Montoya AM, Luna-Rodríguez CE, Treviño-Rangel RDJ, Becerril-García M, Ballesteros-Elizondo RG, Saucedo-Cárdenas O, González GM. In vivo pathogenicity of Trichosporon asahii isolates with different in vitro enzymatic profiles in an immunocompetent murine model of systemic trichosporonosis. Med Mycol 2019; 56:434-441. [PMID: 28992352 DOI: 10.1093/mmy/myx057] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 07/13/2017] [Indexed: 01/19/2023] Open
Abstract
Trichosporon asahii is an opportunistic yeastlike fungus that colonizes the gastrointestinal and respiratory tracts and human skin. Although it is an important cause of disseminated infections by non-Candida species, there are a few reports related to its virulence factors and their possible role in in vivo pathogenicity. We developed a murine model of disseminated trichosporonosis in immunocompetent mice for the evaluation of the in vivo pathogenicity of 6 T. asahii isolates with different in vitro virulence factor profiles. Tissue fungal burden was determined on days 1, 3, 7, 15, and 25 post-challenge. Overall, the largest fungal load was detected in the kidney on the 5 experimental days, while brain, spleen, and liver displayed a comparatively low fungal count. We observed a fungal burden decrease in most experimental groups from day 15. Histological analysis showed the presence of T. asahii in tissue and a generalized inflammatory infiltrate of polymorphonuclear cells in the kidney, liver, red pulp of the spleen, and the hippocampus. Even though our isolates showed different in vitro virulence factors profiles, we did not detect relevant differences when assayed in vivo, except for a higher persistence of a protease- and biofilm-producing strain in kidney, liver, and brain.
Collapse
Affiliation(s)
- Alexandra M Montoya
- Departamento de Microbiología and Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Carolina E Luna-Rodríguez
- Departamento de Microbiología and Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Rogelio de J Treviño-Rangel
- Departamento de Microbiología and Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Miguel Becerril-García
- Departamento de Microbiología and Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey, México
| | | | - Odila Saucedo-Cárdenas
- Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Gloria M González
- Departamento de Microbiología and Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey, México
| |
Collapse
|