51
|
Buttar J, Kon E, Lee A, Kaur G, Lunken G. Effect of diet on the gut mycobiome and potential implications in inflammatory bowel disease. Gut Microbes 2024; 16:2399360. [PMID: 39287010 PMCID: PMC11409510 DOI: 10.1080/19490976.2024.2399360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 07/31/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024] Open
Abstract
The gut microbiome is a complex, unique entity implicated in the prevention, pathogenesis, and progression of common gastrointestinal diseases. While largely dominated by bacterial populations, advanced sequencing techniques have identified co-inhabiting fungal communities, collectively referred to as the mycobiome. Early studies identified that gut inflammation is associated with altered microbial composition, known as gut dysbiosis. Altered microbial profiles are implicated in various pathological diseases, such as inflammatory bowel disease (IBD), though their role as a cause or consequence of systemic inflammation remains the subject of ongoing research. Diet plays a crucial role in the prevention and management of various diseases and is considered to be an essential regulator of systemic inflammation. This review compiles current literature on the impact of dietary modulation on the mycobiome, showing that dietary changes can alter the fungal architecture of the gut. Further research is required to understand the impact of diet on gut fungi, including the metabolic pathways and enzymes involved in fungal fermentation. Additionally, investigating whether dietary modulation of the gut mycobiome could be utilized as a therapy in IBD is essential.
Collapse
Affiliation(s)
- J Buttar
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - E Kon
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
- BC Children's Hospital Research Institute, Vancouver, Canada
| | - A Lee
- Faculty of Land and Food Systems, University of British Columbia, Vancouver, Canada
| | - G Kaur
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - G Lunken
- Department of Medicine, University of British Columbia, Vancouver, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
- BC Children's Hospital Research Institute, Vancouver, Canada
| |
Collapse
|
52
|
Xiao C, Cao S, Li Y, Luo Y, Liu J, Chen Y, Bai Q, Chen L. Pyroptosis in microbial infectious diseases. Mol Biol Rep 2023; 51:42. [PMID: 38158461 DOI: 10.1007/s11033-023-09078-w] [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: 07/13/2023] [Accepted: 10/30/2023] [Indexed: 01/03/2024]
Abstract
Pyroptosis is a gasdermins-mediated programmed cell death that plays an essential role in immune regulation, and its role in autoimmune disease and cancer has been studied extensively. Increasing evidence shows that various microbial infections can lead to pyroptosis, associated with the occurrence and development of microbial infectious diseases. This study reviews the recent advances in pyroptosis in microbial infection, including bacterial, viral, and fungal infections. We also explore potential therapeutic strategies for treating microbial infection-related diseases by targeting pyroptosis.
Collapse
Affiliation(s)
- Cui Xiao
- Department of Public Health Laboratory Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Saihong Cao
- Department of Public Health Laboratory Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Yiyang Medical College, School of Public Health and Laboratory Medicine, Yiyang, Hunan, 421000, China
| | - Yunfei Li
- Department of Public Health Laboratory Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yuchen Luo
- Department of Public Health Laboratory Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jian Liu
- Department of Public Health Laboratory Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yuyu Chen
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University Infection-Associated Hemophagocytic Syndrome, Changsha, Hunan, 421000, China
| | - Qinqin Bai
- Department of Public Health Laboratory Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Lili Chen
- Department of Public Health Laboratory Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
53
|
Zhang Z, Zhang Y, Zhang M, Yu C, Yang P, Xu M, Ling J, Wu Y, Zhu Z, Chen Y, Shi A, Liu X, Zhang J, Yu P, Zhang D. Food-derived peptides as novel therapeutic strategies for NLRP3 inflammasome-related diseases: a systematic review. Crit Rev Food Sci Nutr 2023; 65:1433-1464. [PMID: 38153262 DOI: 10.1080/10408398.2023.2294164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3), a member of the nucleotide-binding domain (NOD) and leucine-rich repeat sequence (LRR) protein (NLR) family, plays an essential role in the inflammation initiation and inflammatory mediator secretion, and thus is also associated with many disease progressions. Food-derived bioactive peptides (FDBP) exhibit excellent anti-inflammatory activity in both in vivo and in vitro models. They are encrypted in plant, meat, and milk proteins and can be released under enzymatic hydrolysis or fermentation conditions, thereby hindering the progression of hyperuricemia, inflammatory bowel disease, chronic liver disease, neurological disorders, lung injury and periodontitis by inactivating the NLRP3. However, there is a lack of systematic review around FDBP, NLRP3, and NLRP3-related diseases. Therefore, this review summarized FDBP that exert inhibiting effects on NLRP3 inflammasome from different protein sources and detailed their preparation and purification methods. Additionally, this paper also compiled the possible inhibitory mechanisms of FDBP on NLRP3 inflammasomes and its regulatory role in NLRP3 inflammasome-related diseases. Finally, the progress of cutting-edge technologies, including nanoparticle, computer-aided screening strategy and recombinant DNA technology, in the acquisition or encapsulation of NLRP3 inhibitory FDBP was discussed. This review provides a scientific basis for understanding the anti-inflammatory mechanism of FDBP through the regulation of the NLRP3 inflammasome and also provides guidance for the development of therapeutic adjuvants or functional foods enriched with these FDBP.
Collapse
Affiliation(s)
- Ziqi Zhang
- The Second Clinical Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Jiangxi, China
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yuan Zhang
- School of Public Health, Nanchang University, Jiangxi, China
| | - Meiying Zhang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Chenfeng Yu
- Huankui College, Nanchang University, Jiangxi, China
| | - Pingping Yang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Minxuan Xu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Jitao Ling
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Yuting Wu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Zicheng Zhu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yixuan Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ao Shi
- School of Medicine, St. George University of London, London, UK
| | - Xiao Liu
- Cardiology Department, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Deju Zhang
- The Second Clinical Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Jiangxi, China
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Hong Kong
| |
Collapse
|
54
|
Case NT, Westman J, Hallett MT, Plumb J, Farheen A, Maxson ME, MacAlpine J, Liston SD, Hube B, Robbins N, Whitesell L, Grinstein S, Cowen LE. Respiration supports intraphagosomal filamentation and escape of Candida albicans from macrophages. mBio 2023; 14:e0274523. [PMID: 38038475 PMCID: PMC10746240 DOI: 10.1128/mbio.02745-23] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 10/16/2023] [Indexed: 12/02/2023] Open
Abstract
IMPORTANCE Candida albicans is a leading human fungal pathogen that often causes life-threatening infections in immunocompromised individuals. The ability of C. albicans to transition between yeast and filamentous forms is key to its virulence, and this occurs in response to many host-relevant cues, including engulfment by host macrophages. While previous efforts identified C. albicans genes required for filamentation in other conditions, the genes important for this morphological transition upon internalization by macrophages remained largely enigmatic. Here, we employed a functional genomic approach to identify genes that enable C. albicans filamentation within macrophages and uncovered a role for the mitochondrial ribosome, respiration, and the SNF1 AMP-activated kinase complex. Additionally, we showed that glucose uptake and glycolysis by macrophages support C. albicans filamentation. This work provides insights into the metabolic dueling that occurs during the interaction of C. albicans with macrophages and identifies vulnerabilities in C. albicans that could serve as promising therapeutic targets.
Collapse
Affiliation(s)
- Nicola T. Case
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Johannes Westman
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Jonathan Plumb
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Aiman Farheen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Michelle E. Maxson
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jessie MacAlpine
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Sean D. Liston
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Luke Whitesell
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Sergio Grinstein
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Center of the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
55
|
Olivier FAB, Traven A. Quantitative live-cell imaging of Candida albicans escape from immune phagocytes. STAR Protoc 2023; 4:102737. [PMID: 37980567 PMCID: PMC10694764 DOI: 10.1016/j.xpro.2023.102737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/04/2023] [Accepted: 11/02/2023] [Indexed: 11/21/2023] Open
Abstract
Population-level dynamics of host-pathogen interactions can be characterized using quantitative live-cell imaging. Here, we present a protocol for infecting macrophages with the fungal pathogen Candida albicans in vitro and quantitative live-cell imaging of immune and pathogen responses. We describe steps for detailed image analysis and provide resources for quantification of phagocytosis and pathogen escape, as well as macrophage membrane permeabilization and viability. This protocol is modifiable for applications with a range of pathogens, immune cell types, and host-pathogen mechanisms. For complete details on the use and execution of this protocol, please refer to Olivier et al.1.
Collapse
Affiliation(s)
- Françios A B Olivier
- Infection Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Centre to Impact AMR, Monash University, Clayton, VIC 3800, Australia.
| | - Ana Traven
- Infection Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Centre to Impact AMR, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
56
|
Elalouf A, Elalouf H, Rosenfeld A. Modulatory immune responses in fungal infection associated with organ transplant - advancements, management, and challenges. Front Immunol 2023; 14:1292625. [PMID: 38143753 PMCID: PMC10748506 DOI: 10.3389/fimmu.2023.1292625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/20/2023] [Indexed: 12/26/2023] Open
Abstract
Organ transplantation stands as a pivotal achievement in modern medicine, offering hope to individuals with end-stage organ diseases. Advancements in immunology led to improved organ transplant survival through the development of immunosuppressants, but this heightened susceptibility to fungal infections with nonspecific symptoms in recipients. This review aims to establish an intricate balance between immune responses and fungal infections in organ transplant recipients. It explores the fundamental immune mechanisms, recent advances in immune response dynamics, and strategies for immune modulation, encompassing responses to fungal infections, immunomodulatory approaches, diagnostics, treatment challenges, and management. Early diagnosis of fungal infections in transplant patients is emphasized with the understanding that innate immune responses could potentially reduce immunosuppression and promise efficient and safe immuno-modulating treatments. Advances in fungal research and genetic influences on immune-fungal interactions are underscored, as well as the potential of single-cell technologies integrated with machine learning for biomarker discovery. This review provides a snapshot of the complex interplay between immune responses and fungal infections in organ transplantation and underscores key research directions.
Collapse
Affiliation(s)
- Amir Elalouf
- Department of Management, Bar-Ilan University, Ramat Gan, Israel
| | - Hadas Elalouf
- Information Science Department, Bar-Ilan University, Ramat Gan, Israel
| | - Ariel Rosenfeld
- Information Science Department, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
57
|
Li S, Liu Y, Weng L, Zhao Y, Zhang Y, Zhang Z, Yang Y, Chen Q, Liu X, Zhang H. The F 1F o-ATP synthase α subunit of Candida albicans induces inflammatory responses by controlling amino acid catabolism. Virulence 2023; 14:2190645. [PMID: 36914568 PMCID: PMC10072111 DOI: 10.1080/21505594.2023.2190645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 02/08/2023] [Accepted: 03/09/2023] [Indexed: 03/15/2023] Open
Abstract
Sepsis is a leading cause of fatality in invasive candidiasis. The magnitude of the inflammatory response is a determinant of sepsis outcomes, and inflammatory cytokine imbalances are central to the pathophysiological processes. We previously demonstrated that a Candida albicans F1Fo-ATP synthase α subunit deletion mutant was nonlethal to mice. Here, the potential effects of the F1Fo-ATP synthase α subunit on host inflammatory responses and the mechanism were studied. Compared with wild-type strain, the F1Fo-ATP synthase α subunit deletion mutant failed to induce inflammatory responses in Galleria mellonella and murine systemic candidiasis models and significantly decreased the mRNA levels of the proinflammatory cytokines IL-1β, IL-6 and increased those of the anti-inflammatory cytokine IL-4 in the kidney. During C. albicans-macrophage co-culture, the F1Fo-ATP synthase α subunit deletion mutant was trapped inside macrophages in yeast form, and its filamentation, a key factor in inducing inflammatory responses, was inhibited. In the macrophage-mimicking microenvironment, the F1Fo-ATP synthase α subunit deletion mutant blocked the cAMP/PKA pathway, the core filamentation-regulating pathway, because it failed to alkalinize environment by catabolizing amino acids, an important alternative carbon source inside macrophages. The mutant downregulated Put1 and Put2, two essential amino acid catabolic enzymes, possibly due to severely impaired oxidative phosphorylation. Our findings reveal that the C. albicans F1Fo-ATP synthase α subunit induces host inflammatory responses by controlling its own amino acid catabolism and it is significant to find drugs that inhibit F1Fo-ATP synthase α subunit activity to control the induction of host inflammatory responses.
Collapse
Affiliation(s)
- Shuixiu Li
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Institute of Mycology, Jinan University, Guangzhou, Guangdong, China
| | - Yuting Liu
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Institute of Mycology, Jinan University, Guangzhou, Guangdong, China
| | - Luobei Weng
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Institute of Mycology, Jinan University, Guangzhou, Guangdong, China
| | - Yajing Zhao
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Institute of Mycology, Jinan University, Guangzhou, Guangdong, China
| | - Yishan Zhang
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Institute of Mycology, Jinan University, Guangzhou, Guangdong, China
| | - Zhanpeng Zhang
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Institute of Mycology, Jinan University, Guangzhou, Guangdong, China
| | - Yang Yang
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Institute of Mycology, Jinan University, Guangzhou, Guangdong, China
| | - Qiaoxin Chen
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Institute of Mycology, Jinan University, Guangzhou, Guangdong, China
| | - Xiaocong Liu
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Institute of Mycology, Jinan University, Guangzhou, Guangdong, China
| | - Hong Zhang
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Institute of Mycology, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
58
|
Mori T, Kataoka H, Into T. Effect of NLRP3 deficiency on cytotoxic and IL-1β-producing activities of synthetic candidalysin peptide. J Oral Biosci 2023; 65:287-292. [PMID: 37659475 DOI: 10.1016/j.job.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/24/2023] [Accepted: 08/27/2023] [Indexed: 09/04/2023]
Abstract
OBJECTIVES Candidalysin (CL), a hydrophobic peptide toxin secreted by Candida albicans, is a key virulence factor that contributes to cytolysis, tissue damage, and immune activation. CL is thought to exert some of its biological activities, including IL-1β production, through the activation of the NLRP3-inflammasome pathway. To date, the mechanism by which CL affects human NLRP3 is not fully understood. We investigated specific activities of synthetic CL peptides using human-derived NLRP3-deficient cells. METHODS Two distinct synthetic CL peptide solutions were prepared: CLd, with CL completely solubilized as nanoparticles in dimethyl sulfoxide, and CLw, with CL partly solubilized in water, and including insoluble microparticles. THP-1 human monocytic cells and NLRP3-deficient THP-1 cells were differentiated into macrophages and stimulated with these peptide solutions. Cell membrane damage, lactate dehydrogenase release, IL-1β production, and caspase-1 activation in stimulated cells were subsequently evaluated. RESULTS Both CLd and CLw exhibited cytotoxic activities independent of NLRP3. Importantly, CLd induced IL-1β production and caspase-1 activation in an NLRP3-independent manner, whereas these activities in CLw-stimulated cells were entirely NLRP3-dependent, suggesting that the NLRP3-dependent response might be triggered by insoluble microparticles. CONCLUSIONS Our results demonstrate that inherent CL activities can cause cell damage and IL-1β production in an NLRP3-independent manner. Our research advances the elucidation of the role of NLRP3 in CL biological activity, underscoring the necessity for further exploration of the precise mechanisms underlying the NLRP3-independent effects of CL and providing novel insights into the complexity of host-pathogen interactions.
Collapse
Affiliation(s)
- Taiki Mori
- Department of Oral Microbiology, Division of Oral Infections Health Sciences, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu 501-0296, Japan
| | - Hideo Kataoka
- Department of Oral Microbiology, Division of Oral Infections Health Sciences, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu 501-0296, Japan
| | - Takeshi Into
- Department of Oral Microbiology, Division of Oral Infections Health Sciences, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu 501-0296, Japan.
| |
Collapse
|
59
|
Miramón P, Pountain AW, Lorenz MC. Candida auris-macrophage cellular interactions and transcriptional response. Infect Immun 2023; 91:e0027423. [PMID: 37815367 PMCID: PMC10652981 DOI: 10.1128/iai.00274-23] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 08/29/2023] [Indexed: 10/11/2023] Open
Abstract
The pathogenic yeast Candida auris represents a global threat of the utmost clinical relevance. This emerging fungal species is remarkable in its resistance to commonly used antifungal agents and its persistence in the nosocomial settings. The innate immune system is one the first lines of defense preventing the dissemination of pathogens in the host. C. auris is susceptible to circulating phagocytes, and understanding the molecular details of these interactions may suggest routes to improved therapies. In this work, we examined the interactions of this yeast with macrophages. We found that macrophages avidly phagocytose C. auris; however, intracellular replication is not inhibited, indicating that C. auris resists the killing mechanisms imposed by the phagocyte. Unlike Candida albicans, phagocytosis of C. auris does not induce macrophage lysis. The transcriptional response of C. auris to macrophage phagocytosis is very similar to other members of the CUG clade (C. albicans, C. tropicalis, C. parapsilosis, C. lusitaniae), i.e., downregulation of transcription/translation and upregulation of alternative carbon metabolism pathways, transporters, and induction of oxidative stress response and proteolysis. Gene family expansions are common in this yeast, and we found that many of these genes are induced in response to macrophage co-incubation. Among these, amino acid and oligopeptide transporters, as well as lipases and proteases, are upregulated. Thus, C. auris shares key transcriptional signatures shared with other fungal pathogens and capitalizes on the expansion of gene families coding for potential virulence attributes that allow its survival, persistence, and evasion of the innate immune system.
Collapse
Affiliation(s)
- Pedro Miramón
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, USA
| | | | - Michael C. Lorenz
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, USA
| |
Collapse
|
60
|
Carlson SL, Mathew L, Savage M, Kok K, Lindsay JO, Munro CA, McCarthy NE. Mucosal Immunity to Gut Fungi in Health and Inflammatory Bowel Disease. J Fungi (Basel) 2023; 9:1105. [PMID: 37998910 PMCID: PMC10672531 DOI: 10.3390/jof9111105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/09/2023] [Accepted: 11/12/2023] [Indexed: 11/25/2023] Open
Abstract
The gut microbiome is a diverse microbial community composed of bacteria, viruses, and fungi that plays a major role in human health and disease. Dysregulation of these gut organisms in a genetically susceptible host is fundamental to the pathogenesis of inflammatory bowel disease (IBD). While bacterial dysbiosis has been a predominant focus of research for many years, there is growing recognition that fungal interactions with the host immune system are an important driver of gut inflammation. Candida albicans is likely the most studied fungus in the context of IBD, being a near universal gut commensal in humans and also a major barrier-invasive pathogen. There is emerging evidence that intra-strain variation in C. albicans virulence factors exerts a critical influence on IBD pathophysiology. In this review, we describe the immunological impacts of variations in C. lbicans colonisation, morphology, genetics, and proteomics in IBD, as well as the clinical and therapeutic implications.
Collapse
Affiliation(s)
- Sean L. Carlson
- Centre for Immunobiology, The Blizard Institute, Queen Mary University of London, London E1 2AT, UK
- Gastroenterology Department, Royal London Hospital, Barts Health NHS Trust, London E1 1BB, UK
| | - Liya Mathew
- Centre for Immunobiology, The Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Michael Savage
- Centre for Immunobiology, The Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Klaartje Kok
- Centre for Immunobiology, The Blizard Institute, Queen Mary University of London, London E1 2AT, UK
- Gastroenterology Department, Royal London Hospital, Barts Health NHS Trust, London E1 1BB, UK
| | - James O. Lindsay
- Centre for Immunobiology, The Blizard Institute, Queen Mary University of London, London E1 2AT, UK
- Gastroenterology Department, Royal London Hospital, Barts Health NHS Trust, London E1 1BB, UK
| | - Carol A. Munro
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB24 3FX, UK
| | - Neil E. McCarthy
- Centre for Immunobiology, The Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| |
Collapse
|
61
|
Unger L, Skoluda S, Backman E, Amulic B, Ponce‐Garcia FM, Etiaba CNC, Yellagunda S, Krüger R, von Bernuth H, Bylund J, Hube B, Naglik JR, Urban CF. Candida albicans induces neutrophil extracellular traps and leucotoxic hypercitrullination via candidalysin. EMBO Rep 2023; 24:e57571. [PMID: 37795769 PMCID: PMC10626426 DOI: 10.15252/embr.202357571] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/05/2023] [Accepted: 09/18/2023] [Indexed: 10/06/2023] Open
Abstract
The peptide toxin candidalysin, secreted by Candida albicans hyphae, promotes stimulation of neutrophil extracellular traps (NETs). However, candidalysin alone triggers a distinct mechanism for NET-like structures (NLS), which are more compact and less fibrous than canonical NETs. Candidalysin activates NADPH oxidase and calcium influx, with both processes contributing to morphological changes in neutrophils resulting in NLS formation. NLS are induced by leucotoxic hypercitrullination, which is governed by calcium-induced protein arginine deaminase 4 activation and initiation of intracellular signalling events in a dose- and time-dependent manner. However, activation of signalling by candidalysin does not suffice to trigger downstream events essential for NET formation, as demonstrated by lack of lamin A/C phosphorylation, an event required for activation of cyclin-dependent kinases that are crucial for NET release. Candidalysin-triggered NLS demonstrate anti-Candida activity, which is resistant to nuclease treatment and dependent on the deprivation of Zn2+ . This study reveals that C. albicans hyphae releasing candidalysin concurrently trigger canonical NETs and NLS, which together form a fibrous sticky network that entangles C. albicans hyphae and efficiently inhibits their growth.
Collapse
Affiliation(s)
- Lucas Unger
- Department of Clinical MicrobiologyUmeå UniversityUmeåSweden
- Umeå Centre for Microbial Research (UCMR)Umeå UniversityUmeåSweden
| | - Samuel Skoluda
- Department of Clinical MicrobiologyUmeå UniversityUmeåSweden
- Umeå Centre for Microbial Research (UCMR)Umeå UniversityUmeåSweden
| | - Emelie Backman
- Department of Clinical MicrobiologyUmeå UniversityUmeåSweden
- Umeå Centre for Microbial Research (UCMR)Umeå UniversityUmeåSweden
| | - Borko Amulic
- School of Cellular and Molecular MedicineUniversity of BristolBristolUK
| | | | - Chinelo NC Etiaba
- School of Cellular and Molecular MedicineUniversity of BristolBristolUK
| | - Sujan Yellagunda
- Department of Clinical MicrobiologyUmeå UniversityUmeåSweden
- Umeå Centre for Microbial Research (UCMR)Umeå UniversityUmeåSweden
| | - Renate Krüger
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care MedicineCharité – Universitätsmedizin BerlinBerlinGermany
| | - Horst von Bernuth
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care MedicineCharité – Universitätsmedizin BerlinBerlinGermany
- Department of ImmunologyLabor Berlin Labor Berlin – Charité Vivantes GmbHBerlinGermany
- Berlin Institute of Health at Charité – Universitätsmedizin BerlinBerlinGermany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of Health (BIH)Berlin‐Brandenburg Center for Regenerative Therapies (BCRT)BerlinGermany
| | - Johan Bylund
- Department of Oral Microbiology & Immunology, Institute of OdontologySahlgrenska Academy at University of GothenburgGothenburgSweden
| | - Bernhard Hube
- Department of Microbial Pathogenicity MechanismsLeibniz Institute for Natural Product Research and Infection Biology ‐ Hans‐Knoell‐InstituteJenaGermany
- Friedrich Schiller UniversityJenaGermany
| | - Julian R Naglik
- Centre for Host‐Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial SciencesKing's College LondonLondonUK
| | - Constantin F Urban
- Department of Clinical MicrobiologyUmeå UniversityUmeåSweden
- Umeå Centre for Microbial Research (UCMR)Umeå UniversityUmeåSweden
| |
Collapse
|
62
|
Deng R, Wang X, Li R. Dermatophyte infection: from fungal pathogenicity to host immune responses. Front Immunol 2023; 14:1285887. [PMID: 38022599 PMCID: PMC10652793 DOI: 10.3389/fimmu.2023.1285887] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Dermatophytosis is a common superficial infection caused by dermatophytes, a group of pathogenic keratinophilic fungi. Apart from invasion against skin barrier, host immune responses to dermatophytes could also lead to pathologic inflammation and tissue damage to some extent. Therefore, it is of great help to understand the pathogenesis of dermatophytes, including fungal virulence factors and anti-pathogen immune responses. This review aims to summarize the recent advances in host-fungal interactions, focusing on the mechanisms of anti-fungal immunity and the relationship between immune deficiency and chronic dermatophytosis, in order to facilitate novel diagnostic and therapeutic approaches to improve the outcomes of these patients.
Collapse
Affiliation(s)
- Ruixin Deng
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China
- Research Center for Medical Mycology, Peking University, Beijing, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China
| | - Xiaowen Wang
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China
- Research Center for Medical Mycology, Peking University, Beijing, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China
| | - Ruoyu Li
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China
- Research Center for Medical Mycology, Peking University, Beijing, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China
| |
Collapse
|
63
|
Hsu CL, Schnabl B. The gut-liver axis and gut microbiota in health and liver disease. Nat Rev Microbiol 2023; 21:719-733. [PMID: 37316582 PMCID: PMC10794111 DOI: 10.1038/s41579-023-00904-3] [Citation(s) in RCA: 200] [Impact Index Per Article: 100.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2023] [Indexed: 06/16/2023]
Abstract
The trillions of microorganisms in the human intestine are important regulators of health, and disruptions in the gut microbial communities can cause disease. The gut, liver and immune system have a symbiotic relationship with these microorganisms. Environmental factors, such as high-fat diets and alcohol consumption, can disrupt and alter microbial communities. This dysbiosis can lead to dysfunction of the intestinal barrier, translocation of microbial components to the liver and development or progression of liver disease. Changes in metabolites produced by gut microorganisms can also contribute to liver disease. In this Review, we discuss the importance of the gut microbiota in maintenance of health and the alterations in microbial mediators that contribute to liver disease. We present strategies for modulation of the intestinal microbiota and/or their metabolites as potential treatments for liver disease.
Collapse
Affiliation(s)
- Cynthia L Hsu
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
- Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA.
| |
Collapse
|
64
|
Xiao Z, Li Y, Xiong L, Liao J, Gao Y, Luo Y, Wang Y, Chen T, Yu D, Wang T, Zhang C, Chen Z. Recent Advances in Anti-Atherosclerosis and Potential Therapeutic Targets for Nanomaterial-Derived Drug Formulations. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302918. [PMID: 37698552 PMCID: PMC10582432 DOI: 10.1002/advs.202302918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/12/2023] [Indexed: 09/13/2023]
Abstract
Atherosclerosis, the leading cause of death worldwide, is responsible for ≈17.6 million deaths globally each year. Most therapeutic drugs for atherosclerosis have low delivery efficiencies and significant side effects, and this has hampered the development of effective treatment strategies. Diversified nanomaterials can improve drug properties and are considered to be key for the development of improved treatment strategies for atherosclerosis. The pathological mechanisms underlying atherosclerosis is summarized, rationally designed nanoparticle-mediated therapeutic strategies, and potential future therapeutic targets for nanodelivery. The content of this study reveals the potential and challenges of nanoparticle use for the treatment of atherosclerosis and highlights new effective design ideas.
Collapse
Affiliation(s)
- Zhicheng Xiao
- Shanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Yi Li
- Shanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Liyan Xiong
- Shanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Jun Liao
- Shanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Yijun Gao
- Shanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Yunchun Luo
- Shanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Yun Wang
- Shanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Ting Chen
- Shanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Dahai Yu
- Weihai Medical Area970 Hospital of Joint Logistic Support Force of PLAWeihai264200China
| | - Tingfang Wang
- Shanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Chuan Zhang
- Shanghai Engineering Research Center of Organ RepairSchool of MedicineShanghai UniversityShanghai200444China
| | - Zhe‐Sheng Chen
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's UniversityNew York11439USA
| |
Collapse
|
65
|
Liang X, Chen D, Wang J, Liao B, Shen J, Ye X, Wang Z, Zhu C, Gou L, Zhou X, Cheng L, Ren B, Zhou X. Artemisinins inhibit oral candidiasis caused by Candida albicans through the repression on its hyphal development. Int J Oral Sci 2023; 15:40. [PMID: 37699886 PMCID: PMC10497628 DOI: 10.1038/s41368-023-00245-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/14/2023] Open
Abstract
Candida albicans is the most abundant fungal species in oral cavity. As a smart opportunistic pathogen, it increases the virulence by switching its forms from yeasts to hyphae and becomes the major pathogenic agent for oral candidiasis. However, the overuse of current clinical antifungals and lack of new types of drugs highlight the challenges in the antifungal treatments because of the drug resistance and side effects. Anti-virulence strategy is proved as a practical way to develop new types of anti-infective drugs. Here, seven artemisinins, including artemisinin, dihydroartemisinin, artemisinic acid, dihydroartemisinic acid, artesunate, artemether and arteether, were employed to target at the hyphal development, the most important virulence factor of C. albicans. Artemisinins failed to affect the growth, but significantly inhibited the hyphal development of C. albicans, including the clinical azole resistant isolates, and reduced their damage to oral epithelial cells, while arteether showed the strongest activities. The transcriptome suggested that arteether could affect the energy metabolism of C. albicans. Seven artemisinins were then proved to significantly inhibit the productions of ATP and cAMP, while reduced the hyphal inhibition on RAS1 overexpression strain indicating that artemisinins regulated the Ras1-cAMP-Efg1 pathway to inhibit the hyphal development. Importantly, arteether significantly inhibited the fungal burden and infections with no systemic toxicity in the murine oropharyngeal candidiasis models in vivo caused by both fluconazole sensitive and resistant strains. Our results for the first time indicated that artemisinins can be potential antifungal compounds against C. albicans infections by targeting at its hyphal development.
Collapse
Affiliation(s)
- Xiaoyue Liang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ding Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiannan Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Binyou Liao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiawei Shen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xingchen Ye
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zheng Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chengguang Zhu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lichen Gou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xinxuan Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Biao Ren
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China.
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
66
|
Lin WY, Iliev ID. Gut epithelium modulates fungal pathogenesis. Science 2023; 381:483-484. [PMID: 37535732 PMCID: PMC10790205 DOI: 10.1126/science.adj1240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
Specialized epithelium secretes an antifungal peptide.
Collapse
Affiliation(s)
- Woan-Yu Lin
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Iliyan D Iliev
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| |
Collapse
|
67
|
Lu H, Hong T, Jiang Y, Whiteway M, Zhang S. Candidiasis: From cutaneous to systemic, new perspectives of potential targets and therapeutic strategies. Adv Drug Deliv Rev 2023; 199:114960. [PMID: 37307922 DOI: 10.1016/j.addr.2023.114960] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/14/2023]
Abstract
Candidiasis is an infection caused by fungi from a Candida species, most commonly Candida albicans. C. albicans is an opportunistic fungal pathogen typically residing on human skin and mucous membranes of the mouth, intestines or vagina. It can cause a wide variety of mucocutaneous barrier and systemic infections; and becomes a severe health problem in HIV/AIDS patients and in individuals who are immunocompromised following chemotherapy, treatment with immunosuppressive agents or after antibiotic-induced dysbiosis. However, the immune mechanism of host resistance to C. albicans infection is not fully understood, there are a limited number of therapeutic antifungal drugs for candidiasis, and these have disadvantages that limit their clinical application. Therefore, it is urgent to uncover the immune mechanisms of the host protecting against candidiasis and to develop new antifungal strategies. This review synthesizes current knowledge of host immune defense mechanisms from cutaneous candidiasis to invasive C. albicans infection and documents promising insights for treating candidiasis through inhibitors of potential antifungal target proteins.
Collapse
Affiliation(s)
- Hui Lu
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Ting Hong
- Department of Anesthesiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Yuanying Jiang
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Malcolm Whiteway
- Department of Biology, Concordia University, Montreal, QC, Canada.
| | - Shiqun Zhang
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China.
| |
Collapse
|
68
|
Chen HT, Li JS, Li J, Li L, Xu ZC, Zhang Y, Wang RR. Lactobacillus murinus: A key factor in suppression of enterogenous Candida albicans infections in Compound Agrimony enteritis capsules-treated mice. JOURNAL OF ETHNOPHARMACOLOGY 2023; 311:116361. [PMID: 36963475 DOI: 10.1016/j.jep.2023.116361] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/04/2023] [Accepted: 03/04/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Compound Agrimony (FuFangXianHeCao, FFXHC) Enteritis Capsules is an ethnomedicine that is derived from Yi Nationality Herbal Medicine for enteritis treatment. We found that FFXHC reduced the mortality outcomes in enterogenic Candida albicans infected mice models and increased the abundance of Lactobacillus murinus in the intestines. Lactobacillus murinus exhibited comparable therapeutic effects to those of FFXHC in enterogenic Candida albicans infected mice. This study provides novel perspectives into the pharmacological mechanisms of FFXHC. AIM OF THE STUDY We investigated the mechanisms via which FFXHC inhibits C. albicans infections and its effects on L. murinus. MATERIALS AND METHODS Enterogenous C. albicans infection mice models were established and various parameters, including survival rate, weight change, number of colonies, treatment effects on intestinal mucosa, microecology, and immune cytokines evaluated. Susceptibility of C. albicans to L. murinus was evaluated in vitro. RESULTS Treatment with FFXHC reduced the number of colonies, improved the health status, enhanced the survival rates, increased the abundance of L. murinus, reduced damage to the intestinal mucosa, and elevated occludin as well as claudin-1 levels. Interestingly, TNF-α, IFN-γ, IL-10, IL-22, and IL-17A levels were increased while IL-1β levels were suppressed in the intestinal mucosa without any change in peripheral blood cytokine levels. Moreover, FFXHC promoted L. murinus proliferation. This study also confirmed the incubation-dependent anti-C. albicans effects exerted by the metabolic supernatants of L. murinus. CONCLUSIONS FFXHC effectively alleviated intestinal infections of C. albicans in mice and increased the abundance of L. murinus. Supplementation of L. murinus in food can achieve the effects that are comparable to those of FFXHC. Thus, L. murinus maybe essential in FFXHC-based treatment of intestinal C. albicans infections.
Collapse
Affiliation(s)
- Hui-Ting Chen
- Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Jia-Sheng Li
- Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Jun Li
- Yunnan University of Chinese Medicine, Kunming, 650500, China; Hospital of Traditional Chinese Medicine and Western Medicine of Panzhihua, Panzhihua, 617099, China
| | - Li Li
- Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Zhi-Chang Xu
- Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Yi Zhang
- Yunnan University of Chinese Medicine, Kunming, 650500, China.
| | - Rui-Rui Wang
- Yunnan University of Chinese Medicine, Kunming, 650500, China.
| |
Collapse
|
69
|
Li F, Gao Y, Cheng W, Su X, Yang R. Gut fungal mycobiome: A significant factor of tumor occurrence and development. Cancer Lett 2023; 569:216302. [PMID: 37451425 DOI: 10.1016/j.canlet.2023.216302] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/28/2023] [Accepted: 07/01/2023] [Indexed: 07/18/2023]
Abstract
A variety of bacteria, viruses, fungi, protists, archaea and protozoa coexists within the mammalian gastrointestinal (GI) tract such as that fungi are detectable in all intestinal and colon segments in almost all healthy adults. Although fungi can cause infectious diseases, they are also related to gut and systemic homeostasis. Importantly, through transformation of different forms such as from yeast to hyphae, interaction among gut microbiota such as fungal and bacterial interaction, host factors such as immune and host derived factors, and fungus genetic and epigenetic factors, fungi can be transformed from commensal into pathogenic lifestyles. Recent studies have shown that fungi play a significant role in the occurrence and development of tumors such as colorectal cancer. Indeed, evidences have shown that multiple species of different fungi exist in different tumors. Studies have also demonstrated that fungi are related to the occurrence and development of tumors, and also survival of patients. Here we summarize recent advances in the transformation of fungi from commensal into pathogenic lifestyles, and the effects of gut pathogenic fungi on the occurrence and development of tumors such as colorectal and pancreatic cancers.
Collapse
Affiliation(s)
- Fan Li
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300071, China; Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Yunhuan Gao
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300071, China; Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Wenyue Cheng
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300071, China; Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Xiaomin Su
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300071, China; Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Rongcun Yang
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, 300071, China; Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
70
|
Zhu W, Zhang H, Dong Q, Song H, Zhao L. Dual wave of neutrophil recruitment determines the outcome of C. albicans infection. Front Cell Infect Microbiol 2023; 13:1239593. [PMID: 37492529 PMCID: PMC10364056 DOI: 10.3389/fcimb.2023.1239593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 06/26/2023] [Indexed: 07/27/2023] Open
Abstract
Candida albicans is a ubiquitous fungus that can cause superficial and systemic infections in humans. Neutrophils play a crucial role in controlling C. albicans infections. When C. albicans enters the bloodstream, it tends to get trapped in capillary vessels. However, the behavior of neutrophils in combating capillary-residing fungi has not been fully characterized. In this study, we used transgenic mice and whole mount imaging to investigate the growth of C. albicans and its interaction with innate immune cells in different organs. We observed that C. albicans rapidly grows hyphae within hours of infection. Following intravenous infection, we observed two waves of neutrophil recruitment, both of which significantly contributed to the elimination of the fungi. The first wave of neutrophils was induced by complement activation and could be prevented by C5aR blockade. Interestingly, we discovered that the fungicidal effect in the lungs was independent of adhesion molecules such as Mac-1, LFA-1, and ICAM-1. However, these molecules played a more significant role in the optimal killing of C. albicans in the kidney. Importantly, the initial difference in killing efficiency resulted in significantly reduced survival in knockout mice lacking these adhesion molecules. We identified a second wave of neutrophil recruitment associated with hyphal growth and tissue damage, which was independent of the aforementioned adhesion molecules. Overall, this study elucidates the dual wave of neutrophil recruitment during C. albicans infection and highlights the importance of early fungal clearance for favorable disease outcomes.
Collapse
Affiliation(s)
- Weiwei Zhu
- Department of Cardiovascular Medicine, Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Huifang Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Qiming Dong
- Department of Internal Medicine, Greater Baltimore Medical Center, Towson, MD, United States
| | - Hongyong Song
- Department of Cardiovascular Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Lin Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
71
|
Patil RH, Luptáková D, Havlíček V. Infection metallomics for critical care in the post-COVID era. MASS SPECTROMETRY REVIEWS 2023; 42:1221-1243. [PMID: 34854486 DOI: 10.1002/mas.21755] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/15/2021] [Accepted: 07/15/2021] [Indexed: 06/07/2023]
Abstract
Infection metallomics is a mass spectrometry (MS) platform we established based on the central concept that microbial metallophores are specific, sensitive, noninvasive, and promising biomarkers of invasive infectious diseases. Here we review the in vitro, in vivo, and clinical applications of metallophores from historical and functional perspectives, and identify under-studied and emerging application areas with high diagnostic potential for the post-COVID era. MS with isotope data filtering is fundamental to infection metallomics; it has been used to study the interplay between "frenemies" in hosts and to monitor the dynamic response of the microbiome to antibiotic and antimycotic therapies. During infection in critically ill patients, the hostile environment of the host's body activates secondary bacterial, mycobacterial, and fungal metabolism, leading to the production of metallophores that increase the pathogen's chance of survival in the host. MS can reveal the structures, stability, and threshold concentrations of these metal-containing microbial biomarkers of infection in humans and model organisms, and can discriminate invasive disease from benign colonization based on well-defined thresholds distinguishing proliferation from the colonization steady state.
Collapse
Affiliation(s)
- Rutuja H Patil
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
- Department of Analytical Chemistry, Faculty of Science, Palacký University, Olomouc, Czechia
| | - Dominika Luptáková
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Vladimír Havlíček
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
- Department of Analytical Chemistry, Faculty of Science, Palacký University, Olomouc, Czechia
| |
Collapse
|
72
|
Desai JV, Kumar D, Freiwald T, Chauss D, Johnson MD, Abers MS, Steinbrink JM, Perfect JR, Alexander B, Matzaraki V, Snarr BD, Zarakas MA, Oikonomou V, Silva LM, Shivarathri R, Beltran E, Demontel LN, Wang L, Lim JK, Launder D, Conti HR, Swamydas M, McClain MT, Moutsopoulos NM, Kazemian M, Netea MG, Kumar V, Köhl J, Kemper C, Afzali B, Lionakis MS. C5a-licensed phagocytes drive sterilizing immunity during systemic fungal infection. Cell 2023; 186:2802-2822.e22. [PMID: 37220746 PMCID: PMC10330337 DOI: 10.1016/j.cell.2023.04.031] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 03/10/2023] [Accepted: 04/21/2023] [Indexed: 05/25/2023]
Abstract
Systemic candidiasis is a common, high-mortality, nosocomial fungal infection. Unexpectedly, it has emerged as a complication of anti-complement C5-targeted monoclonal antibody treatment, indicating a critical niche for C5 in antifungal immunity. We identified transcription of complement system genes as the top biological pathway induced in candidemic patients and as predictive of candidemia. Mechanistically, C5a-C5aR1 promoted fungal clearance and host survival in a mouse model of systemic candidiasis by stimulating phagocyte effector function and ERK- and AKT-dependent survival in infected tissues. C5ar1 ablation rewired macrophage metabolism downstream of mTOR, promoting their apoptosis and enhancing mortality through kidney injury. Besides hepatocyte-derived C5, local C5 produced intrinsically by phagocytes provided a key substrate for antifungal protection. Lower serum C5a concentrations or a C5 polymorphism that decreases leukocyte C5 expression correlated independently with poor patient outcomes. Thus, local, phagocyte-derived C5 production licenses phagocyte antimicrobial function and confers innate protection during systemic fungal infection.
Collapse
Affiliation(s)
- Jigar V Desai
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy & Infectious Diseases, NIH, Bethesda, MD, USA
| | - Dhaneshwar Kumar
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA; Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, IN, USA
| | - Tilo Freiwald
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Daniel Chauss
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | | | - Michael S Abers
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy & Infectious Diseases, NIH, Bethesda, MD, USA
| | - Julie M Steinbrink
- Department of Medicine, Division of Infectious Diseases, Duke University, Durham, NC, USA
| | - John R Perfect
- Department of Medicine, Division of Infectious Diseases, Duke University, Durham, NC, USA
| | - Barbara Alexander
- Department of Medicine, Division of Infectious Diseases, Duke University, Durham, NC, USA
| | - Vasiliki Matzaraki
- Department of Genetics, University of Groningen, Groningen, the Netherlands
| | - Brendan D Snarr
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy & Infectious Diseases, NIH, Bethesda, MD, USA
| | - Marissa A Zarakas
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy & Infectious Diseases, NIH, Bethesda, MD, USA
| | - Vasileios Oikonomou
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy & Infectious Diseases, NIH, Bethesda, MD, USA
| | - Lakmali M Silva
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD, USA
| | - Raju Shivarathri
- Center for Discovery & Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Emily Beltran
- Complement and Inflammation Research Section, National Heart Lung and Blood Institute, NIH, Bethesda, MD, USA
| | - Luciana Negro Demontel
- Complement and Inflammation Research Section, National Heart Lung and Blood Institute, NIH, Bethesda, MD, USA
| | - Luopin Wang
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, IN, USA
| | - Jean K Lim
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dylan Launder
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Heather R Conti
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Muthulekha Swamydas
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy & Infectious Diseases, NIH, Bethesda, MD, USA
| | - Micah T McClain
- Department of Medicine, Division of Infectious Diseases, Duke University, Durham, NC, USA
| | - Niki M Moutsopoulos
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD, USA
| | - Majid Kazemian
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, IN, USA
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University, Nijmegen, the Netherlands
| | - Vinod Kumar
- Department of Genetics, University of Groningen, Groningen, the Netherlands; Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University, Nijmegen, the Netherlands
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Claudia Kemper
- Complement and Inflammation Research Section, National Heart Lung and Blood Institute, NIH, Bethesda, MD, USA
| | - Behdad Afzali
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy & Infectious Diseases, NIH, Bethesda, MD, USA.
| |
Collapse
|
73
|
Wu H, Wang L, Wang W, Shao Z, Jia X, Xiao H, Chen J. The secretory Candida effector Sce1 licenses fungal virulence by masking the immunogenic β-1,3-glucan and promoting apoptosis of the host cells. MLIFE 2023; 2:159-177. [PMID: 38817625 PMCID: PMC10989805 DOI: 10.1002/mlf2.12066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/28/2023] [Accepted: 04/05/2023] [Indexed: 06/01/2024]
Abstract
Candida albicans deploys a variety of mechanisms such as morphological switch and elicitor release to promote virulence. However, the intricate interactions between the fungus and the host remain poorly understood, and a comprehensive inventory of fungal virulence factors has yet to be established. In this study, we identified a C. albicans secretory effector protein Sce1, whose induction and secretion are associated with vagina-simulative conditions and chlamydospore formation. Sequence alignment showed that Sce1 belongs to a Pir family in C. albicans, which is conserved across several fungi and primarily characterized as a β-glucan binding protein in the Saccharomyces cerevisiae. Mechanically, Sce1 is primarily localized to the cell wall in a cleaved form as an alkali-labile β-1,3-glucan binding protein and plays a role in masking β-glucan in acidic environments and chlamydospores, a feature that might underline C. albicans' ability to evade host immunity. Further, a cleaved short form of Sce1 protein could be released into extracellular compartments and presented in bone marrow-derived macrophages infected with chlamydospores. This cleaved short form of Sce1 also demonstrated a unique ability to trigger the caspases-8/9-dependent apoptosis in various host cells. Correspondingly, genetic deletion of SCE1 led to dampened vaginal colonization of C. albicans and diminished fungal virulence during systemic infection. The discovery of Sce1 as a versatile virulence effector that executes at various compartments sheds light on the fungus-host interactions and C. albicans pathogenesis.
Collapse
Affiliation(s)
- Hongyu Wu
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghaiChina
| | - Li Wang
- The Center for Microbes, Development and Health, Institut Pasteur of Shanghai, Chinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghaiChina
- Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Wenjuan Wang
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghaiChina
| | - Zhugui Shao
- The Center for Microbes, Development and Health, Institut Pasteur of Shanghai, Chinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghaiChina
- Key Laboratory of Infection and Immunity of Shandong Province and Department of Immunology, School of Biomedical SciencesShandong UniversityJinanChina
| | - Xin‐Ming Jia
- Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Hui Xiao
- The Center for Microbes, Development and Health, Institut Pasteur of Shanghai, Chinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghaiChina
| | - Jiangye Chen
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghaiChina
| |
Collapse
|
74
|
Weerasinghe H, Simm C, Djajawi TM, Tedja I, Lo TL, Simpson DS, Shasha D, Mizrahi N, Olivier FAB, Speir M, Lawlor KE, Ben-Ami R, Traven A. Candida auris uses metabolic strategies to escape and kill macrophages while avoiding robust activation of the NLRP3 inflammasome response. Cell Rep 2023; 42:112522. [PMID: 37204928 DOI: 10.1016/j.celrep.2023.112522] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 03/28/2023] [Accepted: 05/01/2023] [Indexed: 05/21/2023] Open
Abstract
Metabolic adaptations regulate the response of macrophages to infection. The contributions of metabolism to macrophage interactions with the emerging fungal pathogen Candida auris are poorly understood. Here, we show that C. auris-infected macrophages undergo immunometabolic reprogramming and increase glycolysis but fail to activate a strong interleukin (IL)-1β cytokine response or curb C. auris growth. Further analysis shows that C. auris relies on its own metabolic capacity to escape from macrophages and proliferate in vivo. Furthermore, C. auris kills macrophages by triggering host metabolic stress through glucose starvation. However, despite causing macrophage cell death, C. auris does not trigger robust activation of the NLRP3 inflammasome. Consequently, inflammasome-dependent responses remain low throughout infection. Collectively, our findings show that C. auris uses metabolic regulation to eliminate macrophages while remaining immunologically silent to ensure its own survival. Thus, our data suggest that host and pathogen metabolism could represent therapeutic targets for C. auris infections.
Collapse
Affiliation(s)
- Harshini Weerasinghe
- Infection Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Centre to Impact AMR, Monash University, Clayton, VIC 3800, Australia
| | - Claudia Simm
- Infection Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Centre to Impact AMR, Monash University, Clayton, VIC 3800, Australia
| | - Tirta Mario Djajawi
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia
| | - Irma Tedja
- Infection Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Centre to Impact AMR, Monash University, Clayton, VIC 3800, Australia
| | - Tricia L Lo
- Infection Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Centre to Impact AMR, Monash University, Clayton, VIC 3800, Australia
| | - Daniel S Simpson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; The Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - David Shasha
- Infectious Diseases Unit, Tel Aviv Sourasky Medical Centre, Tel Aviv, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Naama Mizrahi
- Infectious Diseases Unit, Tel Aviv Sourasky Medical Centre, Tel Aviv, Israel
| | - Françios A B Olivier
- Infection Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Centre to Impact AMR, Monash University, Clayton, VIC 3800, Australia; The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Mary Speir
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia
| | - Kate E Lawlor
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia
| | - Ronen Ben-Ami
- Infectious Diseases Unit, Tel Aviv Sourasky Medical Centre, Tel Aviv, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ana Traven
- Infection Program and the Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Centre to Impact AMR, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
75
|
Abstract
The microbiome may impact cancer development, progression and treatment responsiveness, but its fungal components remain insufficiently studied in this context. In this review, we highlight accumulating evidence suggesting a possible involvement of commensal and pathogenic fungi in modulation of cancer-related processes. We discuss the mechanisms by which fungi can influence tumour biology, locally by activity exerted within the tumour microenvironment, or remotely through secretion of bioactive metabolites, modulation of host immunity and communications with neighbouring bacterial commensals. We examine prospects of utilising fungi-related molecular signatures in cancer diagnosis, patient stratification and assessment of treatment responsiveness, while highlighting challenges and limitations faced in performing such research. In all, we demonstrate that fungi likely constitute important members of mucosal and tumour-residing microbiomes. Exploration of fungal inter-kingdom interactions with the bacterial microbiome and the host and decoding of their causal impacts on tumour biology may enable their harnessing into cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Aurelia Saftien
- Microbiome and Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Jens Puschhof
- Microbiome and Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Eran Elinav
- Microbiome and Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
76
|
Gaziano R, Sabbatini S, Monari C. The Interplay between Candida albicans, Vaginal Mucosa, Host Immunity and Resident Microbiota in Health and Disease: An Overview and Future Perspectives. Microorganisms 2023; 11:1211. [PMID: 37317186 DOI: 10.3390/microorganisms11051211] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/02/2023] [Accepted: 05/02/2023] [Indexed: 06/16/2023] Open
Abstract
Vulvovaginal candidiasis (VVC), which is primarily caused by Candida albicans, is an infection that affects up to 75% of all reproductive-age women worldwide. Recurrent VVC (RVVC) is defined as >3 episodes per year and affects nearly 8% of women globally. At mucosal sites of the vagina, a delicate and complex balance exists between Candida spp., host immunity and local microbial communities. In fact, both immune response and microbiota composition play a central role in counteracting overgrowth of the fungus and maintaining homeostasis in the host. If this balance is perturbed, the conditions may favor C. albicans overgrowth and the yeast-to-hyphal transition, predisposing the host to VVC. To date, the factors that affect the equilibrium between Candida spp. and the host and drive the transition from C. albicans commensalism to pathogenicity are not yet fully understood. Understanding the host- and fungus-related factors that drive VVC pathogenesis is of paramount importance for the development of adequate therapeutic interventions to combat this common genital infection. This review focuses on the latest advances in the pathogenic mechanisms implicated in the onset of VVC and also discusses novel potential strategies, with a special focus on the use of probiotics and vaginal microbiota transplantation in the treatment and/or prevention of recurrent VVC.
Collapse
Affiliation(s)
- Roberta Gaziano
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Samuele Sabbatini
- Department of Medicine and Surgery, Medical Microbiology Section, University of Perugia, 06132 Perugia, Italy
| | - Claudia Monari
- Department of Medicine and Surgery, Medical Microbiology Section, University of Perugia, 06132 Perugia, Italy
| |
Collapse
|
77
|
An D, Xu W, Ge Y, Ge Y, Zhang L, Zhu Y, Zhang Z, Fan J, Gao R, Jiang L, Huang P, Wang J, Chen X. Protection of Oxygen Glucose Deprivation-Induced Human Brain Vascular Pericyte Injury: Beneficial Effects of Bellidifolin in Cellular Pyroptosis. Neurochem Res 2023:10.1007/s11064-023-03943-7. [PMID: 37127800 DOI: 10.1007/s11064-023-03943-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 01/31/2023] [Accepted: 04/20/2023] [Indexed: 05/03/2023]
Abstract
Pericytes play critical roles in the maintenance of brain vascular homeostasis. However, very little is currently known about how pericytes regulate ischemic stroke-induced brain injury. Inflammation is a key event in the pathobiology of stroke, in which the nod-like receptor protein-3 (NLRP3) inflammasome is involved in, triggering sterile inflammatory responses and pyroptosis. In the current study, an immortalized cell line derived from human brain vascular pericytes (HBVPs) was constructed, and it showed that HBVPs challenged with oxygen glucose deprivation (OGD) displays pronounced cellular excretion of LDH, IL-1β, IL-18 and increased PI positive staining. Mechanistically, upon OGD treatment, NLRP3 forms an inflammasome with its adaptor protein apoptosis-associated speck-like protein, containing a caspase recruitment domain (ASC) and caspase-1, manifested as much more co-stainings of NLRP3, ASC and Caspase-1 in HBVPs, accompanied by the increased protein levels of NLRP3, ASC, caspase-1 as well as the pyroptosis-associated protein gasdermin D (GSDMD). Intriguingly, GSDMD-N shuttled to the mitochondrial membrane triggered by OGD exposure, which promoted massive mitochondria-derived ROS generation. Importantly, the invention value of the specific targets was evaluated by treatment with bellidifolin, a kind of ketone compound derived from Swertia chirayita in traditional Tibetan medicine. It showed that bellidifolin exerts beneficial effects and attenuates the formation of NLRP3/ASC/Caspase-1 complex, thereby impeding GSDMD-N shuttling and resultant ROS generation, protecting against OGD-induced HBVPs pyroptosis. Overall, these findings unravel the potential mechanisms of pericyte injury induced by OGD and indicate that bellidifolin may exert its beneficial effects on pyroptosis, thus providing new therapeutic insights into stroke.
Collapse
Affiliation(s)
- Di An
- Department of Emergency Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Weixiao Xu
- Department of Emergency Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Yingxin Ge
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, 211166, Jiangsu, China
| | - Yaning Ge
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, 211166, Jiangsu, China
| | - Linwei Zhang
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, 211166, Jiangsu, China
| | - Yi Zhu
- Department of Emergency Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Zhongman Zhang
- Department of Emergency Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Junting Fan
- Department of Pharmaceutical Analysis, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Rong Gao
- Department of Hygienic Analysis and Detection, Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Lei Jiang
- Department of Emergency Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Peipei Huang
- Department of Emergency Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Jun Wang
- Key Lab of Modern Toxicology (NJMU), Ministry of Education, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, 211166, Jiangsu, China.
- China International Cooperation Center for Environment and Human Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing, 211166, Jiangsu, China.
| | - Xufeng Chen
- Department of Emergency Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
78
|
Han QQ, Le W. NLRP3 Inflammasome-Mediated Neuroinflammation and Related Mitochondrial Impairment in Parkinson's Disease. Neurosci Bull 2023; 39:832-844. [PMID: 36757612 PMCID: PMC10169990 DOI: 10.1007/s12264-023-01023-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/31/2022] [Indexed: 02/10/2023] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder caused by the loss of dopamine neurons in the substantia nigra and the formation of Lewy bodies, which are mainly composed of alpha-synuclein fibrils. Alpha-synuclein plays a vital role in the neuroinflammation mediated by the nucleotide-binding oligomerization domain-, leucine-rich repeat-, and pyrin domain-containing protein 3 (NLRP3) inflammasome in PD. A better understanding of the NLRP3 inflammasome-mediated neuroinflammation and the related mitochondrial impairment during PD progression may facilitate the development of promising therapies for PD. This review focuses on the molecular mechanisms underlying NLRP3 inflammasome activation, comprising priming and protein complex assembly, as well as the role of mitochondrial impairment and its subsequent inflammatory effects on the progression of neurodegeneration in PD. In addition, the therapeutic strategies targeting the NLRP3 inflammasome for PD treatment are discussed, including the inhibitors of NLRP3 inflammatory pathways, mitochondria-focused treatments, microRNAs, and other therapeutic compounds.
Collapse
Affiliation(s)
- Qiu-Qin Han
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China
- Center for Clinical and Translational Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Weidong Le
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China.
- Center for Clinical and Translational Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China.
| |
Collapse
|
79
|
Hu L, Bai G, Xu Q, Zhao G, Jiang N, Yao H, Liu X, Du Z. Candidalysin amplifies the immune inflammatory response in Candida albicans keratitis through the TREM-1/DAP12 pathway. Int Immunopharmacol 2023; 119:110195. [PMID: 37087869 DOI: 10.1016/j.intimp.2023.110195] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/25/2023]
Abstract
Candidalysin is a fungal peptide toxin secreted by Candida albicans hyphae during invasion into epithelial cells. In Candida albicans-infected mucosa, candidalysin causes epithelial cell damage and activates downstream inflammatory responses, especially the release of inflammatory cytokines. However, the role of candidalysin in Candida albicans corneal keratitis remains unexplored. Moreover, it remains unclear whether candidalysin regulates the inflammatory response through the TREM-1/DAP12 pathway in Candida albicans corneal keratitis. In this study, we determined the expression pattern of TREM-1 in a mouse model of Candida albicans corneal keratitis and investigated the molecular mechanism underlying the inflammatory response regulation by candidalysin. The corneal keratitis model was established in C57BL/6 mice. In the GF9 group, mice were pretreated and then treated with the TREM-1 inhibitor GF9; in the candidalysin group, mice were treated with peptide candidalysin; and in the PD98059 group, mice were pretreated with the ERK inhibitor PD98059. Slit-lamp photography, clinical scoring, PCR, western blotting and immunofluorescence assay were performed to observe disease response and GF9 therapeutic efficacy. Pretreatment with candidalysin or PD98059 was performed before Candida albicans infection. GF9 treatment reduced the expression of TREM-1 and cytokines in the infected mouse cornea, whereas candidalysin treatment increased the expression of TREM-1, p-ERK, and cytokines, and this increase was inhibited by GF9. The candidalysin-induced increment of TREM-1, p-ERK, and cytokines was inhibited by PD98059 pretreatment. These data suggest that candidalysin can initiate inflammatory response in Candida albicans corneal keratitis through the TREM-1/DAP12 pathway and can regulate cytokine expression by enhancing ERK phosphorylation.
Collapse
Affiliation(s)
- Liting Hu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, China
| | - Guitao Bai
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, China; Department of Ophthalmology, Zigong First People's Hospital, 42 Shang Yihao Branch Road, ZiGong 643000, China
| | - Qiang Xu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, China
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, China
| | - Nan Jiang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, China
| | - Hua Yao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, China
| | - Xueqing Liu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, China
| | - Zhaodong Du
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, China.
| |
Collapse
|
80
|
Hartmann P, Schnabl B. Fungal infections and the fungal microbiome in hepatobiliary disorders. J Hepatol 2023; 78:836-851. [PMID: 36565724 PMCID: PMC10033447 DOI: 10.1016/j.jhep.2022.12.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022]
Abstract
Liver and biliary diseases affect more than a billion people worldwide, with high associated morbidity and mortality. The impact of the intestinal bacterial microbiome on liver diseases has been well established. However, the fungal microbiome, or mycobiome, has been overlooked for a long time. Recently, several studies have shed light on the role of the mycobiome in the development and progression of hepatobiliary diseases. In particular, the fungal genus Candida has been found to be involved in the pathogenesis of multiple hepatobiliary conditions. Herein, we compare colonisation and infection, describe mycobiome findings in the healthy state and across the various hepatobiliary conditions, and point toward communalities. We detail how quantitation of immune responses to fungal antigens can be employed to predict disease severity, e.g. using antibodies to Saccharomyces cerevisiae or specific anti-Candida albicans antibodies. We also show how fungal products (e.g. beta-glucans, candidalysin) activate the host's immune system to exacerbate liver and biliary diseases. Finally, we describe how the gut mycobiome can be modulated to ameliorate hepatobiliary conditions.
Collapse
Affiliation(s)
- Phillipp Hartmann
- Department of Medicine, University of California San Diego, La Jolla, CA, USA; Department of Pediatrics, University of California San Diego, La Jolla, CA, USA; Division of Gastroenterology, Hepatology & Nutrition, Rady Children's Hospital San Diego, San Diego, CA, USA
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA; Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA.
| |
Collapse
|
81
|
Bao MY, Li M, Bu QR, Yang Y, Song H, Wang CZ, Wang TM, Li N. The effect of herbal medicine in innate immunity to Candida albicans. Front Immunol 2023; 14:1096383. [PMID: 37483621 PMCID: PMC10359817 DOI: 10.3389/fimmu.2023.1096383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 03/06/2023] [Indexed: 07/25/2023] Open
Abstract
Candida albicans (C. albicans) is an opportunistic pathogenic fungus that often causes mucosal and systemic infections. Several pattern recognition receptors (PRRs), such as Toll-like receptors (TLRs) and C-type lectin receptors (CLRs), have been implicated in the host recognition of C. albicans. These PRRs recognize the pathogen-associated molecular patterns (PAMPs) of C. albicans to activate innate immune cells, thereby rapidly inducing various inflammatory responses by activating intracellular signaling cascades. Herbal medicine and its active components deserve priority development due to their low toxicity and high antibacterial, antiviral and antifungal activities. This review discussed the activities of herbal compounds against C. albicans and their related mechanisms, especially their regulatory role on innate immune cells such as neutrophils, macrophages, and dendritic cells (DCs) implicated in C. albicans infections. Our work aims to find new therapeutic drugs and targets to prevent and treat diseases caused by C. albicans infection with the mechanisms by which this fungus interacts with the innate immune response.
Collapse
Affiliation(s)
- Meng-Yuan Bao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Ming Li
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Qing-Ru Bu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Yue Yang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Hang Song
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Chang-Zhong Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Tian-Ming Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Ning Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, China
| |
Collapse
|
82
|
Hsu C, Ghannoum M, Cominelli F, Martino LD. Mycobiome and Inflammatory Bowel Disease: Role in Disease Pathogenesis, Current Approaches and Novel Nutritional-based Therapies. Inflamm Bowel Dis 2023; 29:470-479. [PMID: 35851921 PMCID: PMC9977251 DOI: 10.1093/ibd/izac156] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Indexed: 12/09/2022]
Abstract
Inflammatory bowel disease (IBD), a disorder characterized by chronic inflammation of the gastrointestinal (GI) tract and a range of adverse health effects including diarrhea, abdominal pain, vomiting, and bloody stools, affects nearly 3.1 million genetically susceptible adults in the United States today. Although the etiology of IBD remains unclear, genetics, stress, diet, and gut microbiota dysbiosis- especially in immunocompromised individuals- have been identified as possible causes of disease. Although previous research has largely focused on the role of bacteria in IBD pathogenesis, recently observed alterations of fungal load and biodiversity in the GI tract of afflicted individuals suggest interkingdom interactions amongst different gut microbial communities, particularly between bacteria and fungi. These discoveries point to the potential utilization of treatment approaches such as antibiotics, antifungals, probiotics, and postbiotics that target both bacteria and fungi in managing IBD. In this review, we discuss the impact of specific fungi on disease pathogenesis, with a focus on the highly virulent genus Candida and how the presence of certain co-enzymes impacts its virulence. In addition, we evaluate current gut microbiome-based therapeutic approaches with the intention of better understanding the mechanisms behind novel therapies.
Collapse
Affiliation(s)
- Caitlyn Hsu
- Case Digestive Health Research Institute, Case Western University School of Medicine, Cleveland, Ohio, 44106, USA
| | - Mahmoud Ghannoum
- Center for Medical Mycology and Integrated Microbiome Core, Department of Dermatology, Case Western Reserve University, and University Hospitals Cleveland Medical Center, Cleveland, Ohio, 44106, USA
| | - Fabio Cominelli
- Case Digestive Health Research Institute, Case Western University School of Medicine, Cleveland, Ohio, 44106, USA
- Department of Medicine, Case Western University School of Medicine, Cleveland, Ohio, 44106, USA
- Department of Pathology, Case Western University School of Medicine, Cleveland, Ohio, 44106, USA
| | - Luca Di Martino
- Case Digestive Health Research Institute, Case Western University School of Medicine, Cleveland, Ohio, 44106, USA
- Department of Medicine, Case Western University School of Medicine, Cleveland, Ohio, 44106, USA
| |
Collapse
|
83
|
Lange T, Kasper L, Gresnigt MS, Brunke S, Hube B. "Under Pressure" - How fungi evade, exploit, and modulate cells of the innate immune system. Semin Immunol 2023; 66:101738. [PMID: 36878023 PMCID: PMC10109127 DOI: 10.1016/j.smim.2023.101738] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Indexed: 03/06/2023]
Abstract
The human immune system uses an arsenal of effector mechanisms to prevent and counteract infections. Yet, some fungal species are extremely successful as human pathogens, which can be attributed to a wide variety of strategies by which these fungi evade, exploit, and modulate the immune system. These fungal pathogens normally are either harmless commensals or environmental fungi. In this review we discuss how commensalism, but also life in an environmental niche without human contact, can drive the evolution of diverse and specialized immune evasion mechanisms. Correspondingly, we discuss the mechanisms contributing to the ability of these fungi to cause superficial to life-threatening infections.
Collapse
Affiliation(s)
- Theresa Lange
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany
| | - Lydia Kasper
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany
| | - Mark S Gresnigt
- Junior Research Group Adaptive Pathogenicity Strategies, Hans Knoell Institute, Jena, Germany
| | - Sascha Brunke
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany; Institute of Microbiology, Friedrich Schiller University, Jena, Germany.
| |
Collapse
|
84
|
Garbe E, Thielemann N, Hohner S, Kumar A, Vylkova S, Kurzai O, Martin R. Functional analysis of the Candida albicans ECE1 Promoter. Microbiol Spectr 2023; 11:e0025323. [PMID: 36786567 PMCID: PMC10100963 DOI: 10.1128/spectrum.00253-23] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 01/24/2023] [Indexed: 02/15/2023] Open
Abstract
The formation of hyphae is a key virulence attribute of Candida albicans as they are required for adhesion to and invasion of host cells, and ultimately deep-tissue dissemination. Hyphae also secrete the peptide toxin candidalysin, which is crucial for destruction of host cell membranes. The peptide is derived from a precursor protein encoded by the gene ECE1 which is strongly induced during hyphal growth. Previous studies revealed a very complex regulation of this gene involving several transcription factors. However, the promoter of the gene is still not characterized. Here, we present a functional analysis of the intergenic region upstream of the ECE1 gene. Rapid amplification of cDNA ends (RACE)-PCR was performed to identify the 5' untranslated region, which has a size of 49 bp regardless of the hyphae-inducing condition. By using green fluorescent protein (GFP) reporter constructs we further defined a minimal promoter length of 1,500 bp which was verified by RT-qPCR. Finally, we identified the TATA element required for the expression of the gene. It is located 106 to 109 bp upstream of the ECE1 start codon. Our results illustrate that despite a very short 5' UTR, a relatively long promoter is required to secure ECE1 transcription, indicating a complex regulatory machinery tightly controlling the expression of the gene. IMPORTANCE In recent years it was shown that secretion of the toxic peptide candidalysin from hyphae of the major human fungal pathogen Candida albicans contributes heavily to its virulence. The peptide is derived from a precursor protein which is encoded by the ECE1 gene whose transcription is known to be closely associated with formation of hyphae. Here, we used a GFP reporter system to determine the length of the ECE1 promoter and were able to show that it has a minimal size of 1,500 bp. Surprisingly, the gene has a very short 5' UTR of only 49 bp. In accordance with this, the TATA element required for transcription is located 106 to 109 bp upstream of the start codon. This indicates that ECE1 expression is controlled by a very long promoter allowing a complex network of transcription factors to contribute to the gene's regulation.
Collapse
Affiliation(s)
- Enrico Garbe
- Septomics Research Center, Friedrich Schiller University and Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Nadja Thielemann
- Institute for Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Sina Hohner
- Institute for Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Animesh Kumar
- Institute for Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Slavena Vylkova
- Septomics Research Center, Friedrich Schiller University and Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Oliver Kurzai
- Institute for Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
- Research Group Fungal Septomics, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
- National Reference Center for Invasive Fungal Infections, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Ronny Martin
- Institute for Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
85
|
Ali FE, Ibrahim IM, Ghogar OM, Abd-alhameed EK, Althagafy HS, Hassanein EH. Therapeutic interventions target the NLRP3 inflammasome in ulcerative colitis: Comprehensive study. World J Gastroenterol 2023; 29:1026-1053. [PMID: 36844140 PMCID: PMC9950862 DOI: 10.3748/wjg.v29.i6.1026] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/29/2023] [Accepted: 01/29/2023] [Indexed: 02/10/2023] Open
Abstract
One of the significant health issues in the world is the prevalence of ulcerative colitis (UC). UC is a chronic disorder that mainly affects the colon, beginning with the rectum, and can progress from asymptomatic mild inflammation to extensive inflammation of the entire colon. Understanding the underlying molecular mechanisms of UC pathogenesis emphasizes the need for innovative therapeutic approaches based on identifying molecular targets. Interestingly, in response to cellular injury, the NLR family pyrin domain containing 3 (NLRP3) inflammasome is a crucial part of the inflammation and immunological reaction by promoting caspase-1 activation and the release of interleukin-1β. This review discusses the mechanisms of NLRP3 inflammasome activation by various signals and its regulation and impact on UC.
Collapse
Affiliation(s)
- Fares E.M Ali
- Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| | - Islam M. Ibrahim
- Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| | - Osama M Ghogar
- Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| | - Esraa K. Abd-alhameed
- Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 12345, Egypt
| | - Hanan S. Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah 12345, Saudi Arabia
| | - Emad H.M. Hassanein
- Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| |
Collapse
|
86
|
Brown AJP. Fungal resilience and host-pathogen interactions: Future perspectives and opportunities. Parasite Immunol 2023; 45:e12946. [PMID: 35962618 PMCID: PMC10078341 DOI: 10.1111/pim.12946] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 01/31/2023]
Abstract
We are constantly exposed to the threat of fungal infection. The outcome-clearance, commensalism or infection-depends largely on the ability of our innate immune defences to clear infecting fungal cells versus the success of the fungus in mounting compensatory adaptive responses. As each seeks to gain advantage during these skirmishes, the interactions between host and fungal pathogen are complex and dynamic. Nevertheless, simply compromising the physiological robustness of fungal pathogens reduces their ability to evade antifungal immunity, their virulence, and their tolerance against antifungal therapy. In this article I argue that this physiological robustness is based on a 'Resilience Network' which mechanistically links and controls fungal growth, metabolism, stress resistance and drug tolerance. The elasticity of this network probably underlies the phenotypic variability of fungal isolates and the heterogeneity of individual cells within clonal populations. Consequently, I suggest that the definition of the fungal Resilience Network represents an important goal for the future which offers the clear potential to reveal drug targets that compromise drug tolerance and synergise with current antifungal therapies.
Collapse
Affiliation(s)
- Alistair J P Brown
- Medical Research Council Centre for Medical Mycology at the University of Exeter, Exeter, UK
| |
Collapse
|
87
|
Russell CM, Rybak JA, Miao J, Peters BM, Barrera FN. Candidalysin: Connecting the pore forming mechanism of this virulence factor to its immunostimulatory properties. J Biol Chem 2023; 299:102829. [PMID: 36581211 PMCID: PMC9852700 DOI: 10.1016/j.jbc.2022.102829] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/02/2022] [Accepted: 12/22/2022] [Indexed: 12/27/2022] Open
Abstract
Candida albicans is a deadly pathogen responsible for millions of mucosal and systemic infections per year. The pathobiology of C. albicans is largely dependent on the damaging and immunostimulatory properties of the peptide candidalysin (CL), a key virulence factor. When CL forms pores in the plasma membrane of epithelial cells, it activates a response network grounded in activation of the epidermal growth factor receptor. Prior reviews have characterized the resulting CL immune activation schemas but lacked insights into the molecular mechanism of CL membrane damage. We recently demonstrated that CL functions by undergoing a unique self-assembly process; CL forms polymers and loops in aqueous solution prior to inserting and forming pores in cell membranes. This mechanism, the first of its kind to be observed, informs new therapeutic avenues to treat Candida infections. Recently, variants of CL were identified in other Candida species, providing an opportunity to identify the residues that are key for CL to function. In this review, we connect the ability of CL to damage cell membranes to its immunostimulatory properties.
Collapse
Affiliation(s)
- Charles M Russell
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, USA
| | - Jennifer A Rybak
- School of Genome Science and Technology, University of Tennessee, Knoxville, Tennessee, USA
| | - Jian Miao
- Graduate Program in Pharmaceutical Sciences, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Brian M Peters
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA; Department of Microbiology, Immunology, and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Francisco N Barrera
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, USA.
| |
Collapse
|
88
|
Significance of Pulmonary Endothelial Injury and the Role of Cyclooxygenase-2 and Prostanoid Signaling. BIOENGINEERING (BASEL, SWITZERLAND) 2023; 10:bioengineering10010117. [PMID: 36671689 PMCID: PMC9855370 DOI: 10.3390/bioengineering10010117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023]
Abstract
The endothelium plays a key role in the dynamic balance of hemodynamic, humoral and inflammatory processes in the human body. Its central importance and the resulting therapeutic concepts are the subject of ongoing research efforts and form the basis for the treatment of numerous diseases. The pulmonary endothelium is an essential component for the gas exchange in humans. Pulmonary endothelial dysfunction has serious consequences for the oxygenation and the gas exchange in humans with the potential of consecutive multiple organ failure. Therefore, in this review, the dysfunction of the pulmonary endothel due to viral, bacterial, and fungal infections, ventilator-related injury, and aspiration is presented in a medical context. Selected aspects of the interaction of endothelial cells with primarily alveolar macrophages are reviewed in more detail. Elucidation of underlying causes and mechanisms of damage and repair may lead to new therapeutic approaches. Specific emphasis is placed on the processes leading to the induction of cyclooxygenase-2 and downstream prostanoid-based signaling pathways associated with this enzyme.
Collapse
|
89
|
Panda C, Mahapatra RK. Bi-Directional Relationship Between Autophagy and Inflammasomes in Neurodegenerative Disorders. Cell Mol Neurobiol 2023; 43:115-137. [PMID: 35066716 PMCID: PMC11415217 DOI: 10.1007/s10571-021-01184-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/12/2021] [Indexed: 01/18/2023]
Abstract
The innate immune system, as the first line of cellular defense, triggers a protective response called inflammation when encountered with invading pathogens. Inflammasome is a multi-protein cytosolic signaling complex that induces inflammation and is critical for inflammation-induced pyroptotic cell death. Inflammasome activation has been found associated with neurodegenerative disorders (NDs), inflammatory diseases, and cancer. Autophagy is a crucial intracellular quality control and homeostasis process which removes the dysfunctional organelles, damaged proteins, and pathogens by sequestering the cytosolic components in a double-membrane vesicle, which eventually fuses with lysosome resulting in cargo degradation. Autophagy disruption has been observed in many NDs presented with persistent neuroinflammation and excessive inflammasome activation. An interplay between inflammation activation and the autophagy process has been realized over the last decade. In the case of NDs, autophagy regulates neuroinflammation load and cellular damage either by engulfing the misfolded protein deposits, dysfunctional mitochondria, or the inflammasome complex itself. A healthy two-way regulation between both cellular processes has been realized for cell survival and cell defense during inflammatory conditions. Therefore, clinical interest in the modulation of inflammasome activation by autophagy inducers is rapidly growing. In this review, we discuss the structural basis of inflammasome activation and the mechanistic ideas of the autophagy process in NDs. Along with comments on multiple ways of neuroinflammation regulation by microglial autophagy, we also present a perspective on pharmacological opportunities in this molecular interplay pertaining to NDs.
Collapse
Affiliation(s)
- Chinmaya Panda
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, Odisha, 751024, India
| | - Rajani Kanta Mahapatra
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, Odisha, 751024, India.
| |
Collapse
|
90
|
Candidalysin Is the Hemolytic Factor of Candida albicans. Toxins (Basel) 2022; 14:toxins14120874. [PMID: 36548771 PMCID: PMC9785678 DOI: 10.3390/toxins14120874] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/07/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Candida albicans produces an important virulence factor, the hypha-associated Ece1-derived secreted peptide toxin candidalysin, which is crucial for the establishment of mucosal and systemic infections. C. albicans has also long been known to be hemolytic, yet the hemolytic factor has not been clearly identified. Here, we show that candidalysin is the hemolytic factor of C. albicans. Its hemolytic activity is modulated by fragments of another Ece1 peptide, P7. Hemolysis by candidalysin can be neutralized by the purinergic receptor antagonist pyridoxal-phosphate-6-azophenyl-2',4'-disulfonic acid (PPADS). PPADS also affects candidalysin's ability to intercalate into synthetic membranes. We also describe the neutralization potential of two anti-candidalysin nanobodies, which are promising candidates for future anti-Candida therapy. This work provides evidence that the historically proposed hemolytic factor of C. albicans is in fact candidalysin and sheds more light on the complex roles of this toxin in C. albicans biology and pathogenicity.
Collapse
|
91
|
Lopes JP, Lionakis MS. Pathogenesis and virulence of Candida albicans. Virulence 2022; 13:89-121. [PMID: 34964702 PMCID: PMC9728475 DOI: 10.1080/21505594.2021.2019950] [Citation(s) in RCA: 191] [Impact Index Per Article: 63.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/08/2021] [Accepted: 12/14/2021] [Indexed: 12/18/2022] Open
Abstract
Candida albicans is a commensal yeast fungus of the human oral, gastrointestinal, and genital mucosal surfaces, and skin. Antibiotic-induced dysbiosis, iatrogenic immunosuppression, and/or medical interventions that impair the integrity of the mucocutaneous barrier and/or perturb protective host defense mechanisms enable C. albicans to become an opportunistic pathogen and cause debilitating mucocutaneous disease and/or life-threatening systemic infections. In this review, we synthesize our current knowledge of the tissue-specific determinants of C. albicans pathogenicity and host immune defense mechanisms.
Collapse
Affiliation(s)
- José Pedro Lopes
- From the Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, MD, USA
| | - Michail S. Lionakis
- From the Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, MD, USA
| |
Collapse
|
92
|
Zhang F, Aschenbrenner D, Yoo JY, Zuo T. The gut mycobiome in health, disease, and clinical applications in association with the gut bacterial microbiome assembly. THE LANCET. MICROBE 2022; 3:e969-e983. [PMID: 36182668 DOI: 10.1016/s2666-5247(22)00203-8] [Citation(s) in RCA: 172] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 07/04/2022] [Accepted: 07/11/2022] [Indexed: 02/06/2023]
Abstract
The gut mycobiome (fungi) is a small but crucial component of the gut microbiome in humans. Intestinal fungi regulate host homoeostasis, pathophysiological and physiological processes, and the assembly of the co-residing gut bacterial microbiome. Over the past decade, accumulating studies have characterised the gut mycobiome in health and several pathological conditions. We review the compositional and functional diversity of the gut mycobiome in healthy populations from birth to adulthood. We describe factors influencing the gut mycobiome and the roles of intestinal fungi-especially Candida and Saccharomyces spp-in diseases and therapies with a particular focus on their synergism with the gut bacterial microbiome and host immunity. Finally, we discuss the underappreciated effects of gut fungi in clinical implications, and highlight future microbiome-based therapies that harness the tripartite relationship among the gut mycobiome, bacterial microbiome, and host immunity, aiming to restore a core gut mycobiome and microbiome and to improve clinical efficacy.
Collapse
Affiliation(s)
- Fen Zhang
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, College of Science and Engineering, Jinan University, Guangzhou, China
| | - Dominik Aschenbrenner
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland
| | - Ji Youn Yoo
- College of Nursing, University of Tennessee, Knoxville, TN, USA
| | - Tao Zuo
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yatsen University, Sun Yat-sen University, Guangzhou, China; Laboratory Animals Centre, Zhongshan School of Medicine, Sun Yatsen University, Guangzhou, China.
| |
Collapse
|
93
|
Zhao S, Shang A, Guo M, Shen L, Han Y, Huang X. The advances in the regulation of immune microenvironment by Candida albicans and macrophage cross-talk. Front Microbiol 2022; 13:1029966. [PMID: 36466634 PMCID: PMC9717684 DOI: 10.3389/fmicb.2022.1029966] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 11/07/2022] [Indexed: 04/04/2024] Open
Abstract
Candida albicans (C. albicans) is the most common causative agent of invasive fungal infections in hospitals. The body defends against and eliminates C. albicans infection by various mechanisms of immune response, and the latter mechanism of immune evasion is a major challenge in the clinical management of C. albicans infection. The role of macrophages in combating C. albicans infection has only recently been recognized, but the mechanisms remain to be elucidated. This review focuses on the interaction between C. albicans and macrophages (macrophages), which causes the body to generate an immune response or C. albicans immune escape, and then regulates the body's immune microenvironment, to explore the effect of C. albicans virulence resistance vs. macrophage killing and clarify the role and mechanism of C. albicans pathogenesis. In general, a thorough understanding of the molecular principles driving antifungal drug resistance is essential for the development of innovative treatments that can counteract both existing and emerging fungal threats.
Collapse
Affiliation(s)
- Shuo Zhao
- Department of Dermatology, School of Medicine, Tongji Hospital, Tongji University, Shanghai, China
| | - Anquan Shang
- Department of Laboratory Medicine, The Second People’s Hospital of Lianyungang, Lianyungang, China
| | - Mengchen Guo
- Department of Dermatology, School of Medicine, Tongji Hospital, Tongji University, Shanghai, China
| | - Liangliang Shen
- Department of Dermatology, School of Medicine, Tongji Hospital, Tongji University, Shanghai, China
| | - Yu Han
- Department of Dermatology, School of Medicine, Tongji Hospital, Tongji University, Shanghai, China
| | - Xin Huang
- Department of Dermatology, School of Medicine, Tongji Hospital, Tongji University, Shanghai, China
| |
Collapse
|
94
|
Lenka S, Bhuyan SK, Bhuyan R. Understanding the characteristics of the host genome and microbiome interaction in oral squamous cell carcinoma: a narrative review. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2022. [DOI: 10.1186/s43088-022-00306-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Oral health status is directly associated with microbes present within it. The abundance of microbes at the OSCC site is more than at its control site, representing its possible role in the progression of OSCC development. Dysbiosis of oral microbiota could be a crucial etiological risk factor in the elevation of OSCC. This study aimed to analyze and assess: a) positive regulator microbes of oral cancer and their abundance at the cancer site, b) pathways involved in positive regulator microbes, and c) identification of the most virulent oral oncogenic microbe.
Main body
It is obtained from several studies that microbes belonging to Prevotella, Fusobacterium, Alloprevotella, Capnocytophaga, Porphyromonas, Campylobacter, and Aggregatibacter are detected to be more in number contrast to healthy sites. Fusobacterium nucleatum, Porphyromonas gingivalis, and Candida albicans show molecular pathways linked with OSCC development. Genes encoding for virulent factors like FimA, Gingipains, lipopolysaccharide (P. gingivalis), FadA, Fap2 (F. nucleatum), and zymosan (C. Albicans) are directly involved in elevating oral cancer.
Conclusion
Mostly, the genes that are involved in promoting oral cancer are the genes that generally encode cell wall proteins. The cell wall proteins that is FadA, Fap, and FimA interact with the host's cell and hamper the normal regulation pathway, which leads to activation of cell proliferating pathways, down-regulates apoptotic pathways, cytoskeleton rearrangement, and upregulates the cell cycle checkpoint regulators; as a result, progression of oral cancer occurs.
Collapse
|
95
|
Russell CM, Schaefer KG, Dixson A, Gray ALH, Pyron RJ, Alves DS, Moore N, Conley EA, Schuck RJ, White TA, Do TD, King GM, Barrera FN. The Candida albicans virulence factor candidalysin polymerizes in solution to form membrane pores and damage epithelial cells. eLife 2022; 11:e75490. [PMID: 36173096 PMCID: PMC9522247 DOI: 10.7554/elife.75490] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 08/15/2022] [Indexed: 11/28/2022] Open
Abstract
Candida albicans causes severe invasive candidiasis. C. albicans infection requires the virulence factor candidalysin (CL) which damages target cell membranes. However, the mechanism that CL uses to permeabilize membranes is unclear. We reveal that CL forms membrane pores using a unique mechanism. Unexpectedly, CL readily assembled into polymers in solution. We propose that the basic structural unit in polymer formation is a CL oligomer, which is sequentially added into a string configuration that can close into a loop. CL loops appear to spontaneously insert into the membrane to become pores. A CL mutation (G4W) inhibited the formation of polymers in solution and prevented pore formation in synthetic lipid systems. Epithelial cell studies showed that G4W CL failed to activate the danger response pathway, a hallmark of the pathogenic effect of CL. These results indicate that CL polymerization in solution is a necessary step for the damage of cellular membranes. Analysis of CL pores by atomic force microscopy revealed co-existence of simple depressions and more complex pores, which are likely formed by CL assembled in an alternate oligomer orientation. We propose that this structural rearrangement represents a maturation mechanism that stabilizes pore formation to achieve more robust cellular damage. To summarize, CL uses a previously unknown mechanism to damage membranes, whereby pre-assembly of CL loops in solution leads to formation of membrane pores. Our investigation not only unravels a new paradigm for the formation of membrane pores, but additionally identifies CL polymerization as a novel therapeutic target to treat candidiasis.
Collapse
Affiliation(s)
- Charles M Russell
- Department of Biochemistry & Cellular and Molecular Biology, University of TennesseeKnoxvilleUnited States
| | - Katherine G Schaefer
- Department of Physics and Astronomy, University of MissouriColumbiaUnited States
| | - Andrew Dixson
- Department of Biochemistry & Cellular and Molecular Biology, University of TennesseeKnoxvilleUnited States
| | - Amber LH Gray
- Department of Chemistry, University of TennesseeKnoxvilleUnited States
| | - Robert J Pyron
- Department of Biochemistry & Cellular and Molecular Biology, University of TennesseeKnoxvilleUnited States
| | - Daiane S Alves
- Department of Biochemistry & Cellular and Molecular Biology, University of TennesseeKnoxvilleUnited States
| | - Nicholas Moore
- Department of Biochemistry & Cellular and Molecular Biology, University of TennesseeKnoxvilleUnited States
| | - Elizabeth A Conley
- Department of Physics and Astronomy, University of MissouriColumbiaUnited States
| | - Ryan J Schuck
- Department of Biochemistry & Cellular and Molecular Biology, University of TennesseeKnoxvilleUnited States
| | - Tommi A White
- Department of Biochemistry, University of MissouriColumbiaUnited States
- Electron Microscopy Core, University of MissouriColumbiaUnited States
| | - Thanh D Do
- Department of Chemistry, University of TennesseeKnoxvilleUnited States
| | - Gavin M King
- Department of Physics and Astronomy, University of MissouriColumbiaUnited States
- Department of Biochemistry, University of MissouriColumbiaUnited States
| | - Francisco N Barrera
- Department of Biochemistry & Cellular and Molecular Biology, University of TennesseeKnoxvilleUnited States
| |
Collapse
|
96
|
Altinkaya Çavuş M, Sav H. Opportunistic Candida Infections in Critical COVID-19 Patients. Pol J Microbiol 2022; 71:411-419. [PMID: 36185025 PMCID: PMC9608158 DOI: 10.33073/pjm-2022-036] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/19/2022] [Indexed: 11/24/2022] Open
Abstract
The frequency of opportunistic fungal infections in critically ill patients whose intensive care unit stays are prolonged due to coronavirus disease 2019 (COVID-19) is higher than in the period before COVID-19. We planned this study to improve the management of Candida infections by defining the Candida species, the etiology of infections caused by Candida species, and the antifungal susceptibility of the species. This retrospective study included patients older than 18 hospitalized in the intensive care unit (ICU) with a definitive diagnosis of COVID-19 for seven months (from March 2021 to September 2021). All study data that we recorded in a standard study form were analyzed with TURCOSA (Turcosa Analytics Ltd. Co., Turkey, www.turcosa.com.tr) statistical software. The patients were evaluated in four groups as group 1 (candidemia patients, n = 78), group 2 (candiduria patients, n = 189), group 3 (control patients, n = 57), and group 4 (patients with candidemia in urine cultures taken before Candida was detected in blood culture, n = 42). Candida species were identified using both conventional and VITEK® 2 (BioMérieux, France) methods. The antifungal susceptibility of fungi was determined using the E test method. Of the 5,583 COVID-19 patients followed during the study period, 78 developed candidemia, and 189 developed candiduria. The incidence of candidemia (per 1,000 admissions) was determined to be 1.6. As a result of statistical analysis, we found that Candida albicans was the dominant strain in candidemia and candiduria, and there was no antifungal resistance except for naturally resistant strains. Candida strains grown in blood and urine were the same in 40 of 42 patients. Mortality was 69.2% for group 1, 60.4% for group 2, and 57.8% for group 3. Antifungals were used in 34 (43.5%) patients from group 1, and 95 (50.2%) from group 2. In the candidemia group without antifungal use, mortality was quite high (77.2%). Antifungal use reduced mortality in the group 2 (p < 0.05). Length of ICU stays, comorbidity, broad-spectrum antibiotics, and corticosteroids are independent risk factors for candidemia in critically ill COVID-19 patients. Our study contributes to the knowledge of risk factors for developing COVID-19-related candida infections. The effect of candiduria on the development of candidemia in critically ill COVID-19 patients should be supported by new studies.
Collapse
Affiliation(s)
- Mıne Altinkaya Çavuş
- Department of Intensive Care, University of Health Sciences, Kayseri City Hospital, Kayseri, Turkey, E-mail:
| | - Hafıze Sav
- Department of Mycology, University of Health Sciences, Kayseri City Hospital, Kayseri, Turkey
| |
Collapse
|
97
|
Olivier FAB, Hilsenstein V, Weerasinghe H, Weir A, Hughes S, Crawford S, Vince JE, Hickey MJ, Traven A. The escape of Candida albicans from macrophages is enabled by the fungal toxin candidalysin and two host cell death pathways. Cell Rep 2022; 40:111374. [PMID: 36130496 DOI: 10.1016/j.celrep.2022.111374] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 06/15/2022] [Accepted: 08/26/2022] [Indexed: 11/25/2022] Open
Abstract
The egress of Candida hyphae from macrophages facilitates immune evasion, but it also alerts macrophages to infection and triggers inflammation. To better define the mechanisms, here we develop an imaging assay to directly and dynamically quantify hyphal escape and correlate it to macrophage responses. The assay reveals that Candida escapes by using two pore-forming proteins to permeabilize macrophage membranes: the fungal toxin candidalysin and Nlrp3 inflammasome-activated Gasdermin D. Candidalysin plays a major role in escape, with Nlrp3 and Gasdermin D-dependent and -independent contributions. The inactivation of Nlrp3 does not reduce hyphal escape, and we identify ETosis via macrophage extracellular trap formation as an additional pathway facilitating hyphal escape. Suppressing hyphal escape does not reduce fungal loads, but it does reduce inflammatory activation. Our findings explain how Candida escapes from macrophages by using three strategies: permeabilizing macrophage membranes via candidalysin and engaging two host cell death pathways, Gasdermin D-mediated pyroptosis and ETosis.
Collapse
Affiliation(s)
- Françios A B Olivier
- Infection Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton 3800 VIC, Australia; Centre to Impact AMR, Monash University, Clayton, VIC 3800, Australia
| | | | - Harshini Weerasinghe
- Infection Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton 3800 VIC, Australia; Centre to Impact AMR, Monash University, Clayton, VIC 3800, Australia
| | - Ashley Weir
- The Walter and Eliza Hall Institute of Medical Research, University of Melbourne, Parkville, VIC 3052, Australia; The Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Sebastian Hughes
- The Walter and Eliza Hall Institute of Medical Research, University of Melbourne, Parkville, VIC 3052, Australia; The Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Simon Crawford
- Monash Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, Clayton, VIC 3800, Australia
| | - James E Vince
- The Walter and Eliza Hall Institute of Medical Research, University of Melbourne, Parkville, VIC 3052, Australia; The Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Michael J Hickey
- Monash Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC 3168, Australia
| | - Ana Traven
- Infection Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton 3800 VIC, Australia; Centre to Impact AMR, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
98
|
Solubility affects IL-1β-producing activity of the synthetic candidalysin peptide. PLoS One 2022; 17:e0273663. [PMID: 36040970 PMCID: PMC9426886 DOI: 10.1371/journal.pone.0273663] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/11/2022] [Indexed: 11/25/2022] Open
Abstract
Candidalysin, a peptide toxin produced specifically from hyphae of Candida albicans, plays a crucial role in C. albicans pathogenesis in the oral cavity and vagina. Synthetic peptides have been widely used in previous studies to investigate the bioactivity of candidalysin. Although the solubility of the peptide, which is expected to have a hydrophobic property, has not been well characterized, candidalysin solutions are usually prepared in water. In this study, we prepared the synthetic peptide candidalysin in water (CLw) or in dimethyl sulfoxide (CLd) and compared their cytotoxicity and interleukin (IL)-1β-producing activity to determine whether the activity of the peptide would be affected. In addition, we evaluated whether the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome pathway or other pathways were involved in their activities. Unexpectedly, we found that CLw was not completely solubilized and contained abundant insoluble microparticles. CLw was active at comparably high concentrations (≥ 10 μM). In contrast, CLd is completely solubilized and sufficiently active at low concentrations, that is, 1 μM or less. CLw showed weak cytotoxicity and NLRP3-dependent and cathepsin B-dependent IL-1β-producing activity, whereas CLd showed strong cytotoxicity and cathepsin B-dependent IL-1β-producing activity. Fractionation of CLw revealed that NLRP3-dependent activity was caused by insoluble microparticles. Furthermore, nanoparticle tracking of CLd revealed that the peptide was present as nanoparticles with a size of 96 nm. CLw contained a small amount of such nanoparticles. Thus, the bioactivities of the synthetic peptide candidalysin, especially the IL-1β-producing activity, are affected by the solubility of the peptide depending on the solvent employed. The NLRP3-dependent activity of the synthetic peptide is caused by insoluble microparticles and may not be the intrinsic activity of candidalysin.
Collapse
|
99
|
Liu A, Gao W, Zhu Y, Hou X, Chu H. Gut Non-Bacterial Microbiota: Emerging Link to Irritable Bowel Syndrome. Toxins (Basel) 2022; 14:596. [PMID: 36136534 PMCID: PMC9503233 DOI: 10.3390/toxins14090596] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/18/2022] [Accepted: 08/25/2022] [Indexed: 11/20/2022] Open
Abstract
As a common functional gastrointestinal disorder, irritable bowel syndrome (IBS) significantly affects personal health and imposes a substantial economic burden on society, but the current understanding of its occurrence and treatment is still inadequate. Emerging evidence suggests that IBS is associated with gut microbial dysbiosis, but most studies focus on the bacteria and neglect other communities of the microbiota, including fungi, viruses, archaea, and other parasitic microorganisms. This review summarizes the latest findings that link the nonbacterial microbiota with IBS. IBS patients show less fungal and viral diversity but some alterations in mycobiome, virome, and archaeome, such as an increased abundance of Candida albicans. Moreover, fungi and methanogens can aid in diagnosis. Fungi are related to distinct IBS symptoms and induce immune responses, intestinal barrier disruption, and visceral hypersensitivity via specific receptors, cells, and metabolites. Novel therapeutic methods for IBS include fungicides, inhibitors targeting fungal pathogenic pathways, probiotic fungi, prebiotics, and fecal microbiota transplantation. Additionally, viruses, methanogens, and parasitic microorganisms are also involved in the pathophysiology and treatment. Therefore, the gut nonbacterial microbiota is involved in the pathogenesis of IBS, which provides a novel perspective on the noninvasive diagnosis and precise treatment of this disease.
Collapse
Affiliation(s)
- Ao Liu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Wenkang Gao
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Yixin Zhu
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Xiaohua Hou
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| |
Collapse
|
100
|
Xie Y, Zhou X, Zhang J, Yu H, Song Z. Immunomodulatory responses of differentially polarized macrophages to fungal infections. Int Immunopharmacol 2022; 111:109089. [PMID: 35964406 DOI: 10.1016/j.intimp.2022.109089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/16/2022] [Accepted: 07/22/2022] [Indexed: 11/05/2022]
Abstract
Macrophages, the first line of defense against invasive fungi in the innate immune system, are widely distributed in the blood and tissues of the body. In response to various internal and external stimulators, macrophages can polarize into classically activated macrophages (M1) and alternatively activated macrophages (M2). These two types of polarized macrophages play different roles in antifungal activity and in maintaining the steady-state balance between inflammation and tissue repair. However, the antifungal mechanisms of M1- and M2-type macrophages have not been fully described. In this review, the immune regulatory mechanisms against pathogenic fungi of these two classical types of macrophages in various tissues are summarized. The effects of antifungal factors on macrophage differentiation are also highlighted. The description of these data, on the one hand provides valuable insight for future investigations and also highlights new strategies for the treatment of pathogenic fungal infections.
Collapse
Affiliation(s)
- Yuxin Xie
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, PR China.
| | - Xue Zhou
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, PR China.
| | - Jinping Zhang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, PR China; The Public Platform of Molecular Biotechnology, Public Center of Experimental Technology, Southwest Medical University, Luzhou 646000, People's Republic of China.
| | - Hong Yu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, PR China; The Public Platform of Cell Biotechnology, Public Center of Experimental Technology, Southwest Medical University, Luzhou 646000, PR China.
| | - Zhangyong Song
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, PR China; The Public Platform of Molecular Biotechnology, Public Center of Experimental Technology, Southwest Medical University, Luzhou 646000, People's Republic of China.
| |
Collapse
|