1
|
Wang F, Zhang J, Zhang Q, Song Z, Xin C. Antifungal activities of Equol against Candida albicans in vitro and in vivo. Virulence 2024; 15:2404256. [PMID: 39267283 PMCID: PMC11409501 DOI: 10.1080/21505594.2024.2404256] [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: 01/26/2024] [Revised: 08/25/2024] [Accepted: 09/05/2024] [Indexed: 09/17/2024] Open
Abstract
Candida albicans is an opportunistic fungal pathogen that can cause systemic infections in immunocompromised individuals. Morphological transition and biofilm formation are major virulence factors of C. albicans. Moreover, biofilm enhances resistance to antifungal agents. Therefore, it is urgent to identify new and effective compounds to target the biofilm of C. albicans. In the present study, the antifungal activities of equol against C. albicans were investigated. In vitro, the microdilution analysis and spot assay result showed that equol exhibited potent inhibitory activities against C. albicans. Further investigations confirmed that the antifungal effects of equol involved interference with the transition from yeast to hypha and biofilm formation of C. albicans. In addition, transcriptome sequencing and reverse transcription-quantitative PCR (qRT-PCR) analysis showed that equol significantly downregulated the expression of several genes in the Ras1-cAMP-PKA pathway related to hyphae and biofilm formation and significantly upregulated the expression of the negative transcriptional repressors RFG1 and TUP1. Moreover, equol effectively reduced the production of cAMP, a key messenger in the Ras1-cAMP-PKA pathway, while supplementation with cAMP partly rescued the equol-induced defects in hyphal development. Furthermore, in a mouse model of systemic candidiasis (SC), equol treatment significantly decreased the fungal burden (liver, kidneys, and lung) in mice and local tissue damage, while enhancing the production of interleukin-10 (IL-10). Together, these findings confirm that equol is a potentially effective agent for treatment of SC.
Collapse
Affiliation(s)
- Fen Wang
- Nanobiosensing and Microfluidic Point-of-Care Testing Key Laboratory of LuZhou, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jinping Zhang
- School of Basic Medical Science, Southwest Medical University, Luzhou, People's Republic of China
| | - Qian Zhang
- Department of blood transfusion, Zhejiang people's hospital, Yichang, China
| | - Zhangyong Song
- School of Basic Medical Science, Southwest Medical University, Luzhou, People's Republic of China
- Technical Platform for the Molecular Biology, Research Core Facility, Southwest Medical University, Luzhou, People's Republic of China
- Southwest Medical University, Hemodynamics and Medical Engineering Combination Key Laboratory of Luzhou, Luzhou, People's Republic of China
| | - Caiyan Xin
- School of Basic Medical Science, Southwest Medical University, Luzhou, People's Republic of China
| |
Collapse
|
2
|
Santos LA, Castro Dutra J, Malaquias LCC, Andrade ND, Gomes BN, Burger E. Paracoccidioides spp.: Escape mechanisms and their implications for the development of this mycosis. Microb Pathog 2024; 196:106951. [PMID: 39299555 DOI: 10.1016/j.micpath.2024.106951] [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: 05/28/2024] [Revised: 09/13/2024] [Accepted: 09/15/2024] [Indexed: 09/22/2024]
Abstract
Paracoccidioidomycosis (PCM) is a systemic granulomatous mycosis prevalent in individuals who carry out rural activities. Its etiological agent is a thermodimorphic fungus belonging to the genus; Paracoccidioides spp. Seven species of this fungus are known: Paracoccidioides brasiliensis, Paracoccidioides lutzii, Paracoccidioides americana, Paracoccidioides restrepiensis, Paracoccidioides venezuelensis, Paracoccidioides loboi and Paracoccidioides ceti. For a long time, Paracoccidioides brasiliensis was attributed as the only causal agent of this mycosis. What is known about adhesins, virulence, escape mechanisms and fungal involvement with the host's immune system is correlated with the species Paracoccidioides brasiliensis. Interactions between Paracoccidioides spp. and the host are complex and dynamic. The fungus needs nutrients for its needs and must adapt to a hostile environment, evading the host's immune system, thus enabling the development of the infectious process. On the other hand, the host's immune system recognizes Paracoccidioides spp. and employs all protective mechanisms to prevent fungal growth and consequently tissue invasion. Knowing this, understanding how Paracoccidioides spp. escapes the host's immune system, can help to understand the pathogenic mechanisms related to the development of the disease and, therefore, in the design of new specific treatment strategies. In this review we discuss these mechanisms and what are the adhesion molecules of Paracoccidioides spp. uses to escape the hostile environment imposed by the host's defense mechanisms; finally, we suggest how to neutralize them with new antifungal therapies.
Collapse
Affiliation(s)
- Lauana Aparecida Santos
- Department of Microbiology and Immunology at Federal University of Alfenas - UNIFAL, Alfenas, MG, CEP 37130-001, Brazil
| | - Julia Castro Dutra
- Department of Microbiology and Immunology at Federal University of Alfenas - UNIFAL, Alfenas, MG, CEP 37130-001, Brazil
| | - Luiz Cosme Cotta Malaquias
- Department of Microbiology and Immunology at Federal University of Alfenas - UNIFAL, Alfenas, MG, CEP 37130-001, Brazil
| | - Nayara Dias Andrade
- Department of Microbiology and Immunology at Federal University of Alfenas - UNIFAL, Alfenas, MG, CEP 37130-001, Brazil
| | - Bruno Nascimento Gomes
- Department of Microbiology and Immunology at Federal University of Alfenas - UNIFAL, Alfenas, MG, CEP 37130-001, Brazil
| | - Eva Burger
- Department of Microbiology and Immunology at Federal University of Alfenas - UNIFAL, Alfenas, MG, CEP 37130-001, Brazil.
| |
Collapse
|
3
|
Vernel-Pauillac F, Laurent-Winter C, Fiette L, Janbon G, Aimanianda V, Dromer F. Cryptococcus neoformans infections: aspartyl protease potential to improve outcome in susceptible hosts. mBio 2024:e0273324. [PMID: 39440979 DOI: 10.1128/mbio.02733-24] [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: 09/04/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024] Open
Abstract
Though a confined or a broad population is exposed respectively to endemic or pandemic infections, in the same environment, some individuals resist the development of infections. The attributed reason is the inheritance of a set of immune system genes that can efficiently deal with the pathogens. In this study, we show how outbred mice differentially respond to Cryptococcus neoformans, a fungal pathogen, and the mechanism through which the surviving mice mount a protective immune defense. We identified that those mice developing antibodies specifically against Pep1p, an aspartic protease secreted by C. neoformans, had significantly improved survival. Vaccination (either prophylactic or therapeutic) with a recombinant Pep1p significantly increased the survival of the mice by decreasing the fungal load and stimulating a protective immune response. Passive immunization of C. neoformans-infected mice with monoclonal antibodies developed against Pep1p also improves the survival of the mice by increasing phagocytosis of C. neoformans and decreasing the multiplication of this fungus. Together, these data demonstrate the prophylactic and therapeutic potentials of the C. neoformans antigenic protein Pep1p or Pep1p-specific antibodies against this fungal infection. Also, this study suggests that the immunological interaction and thereby the responses developed against a pathogen guide the hosts to behave differentially against microbial pathogenicity. IMPORTANCE Vaccination and immunotherapies against fungal pathogens still remain a challenge. Here, we show using an in vivo model based on outbred mice that development of antibodies against Pep1p, an antigenic protein of the fungal pathogen Cryptococcus neoformans, confers resistance to this fungal infection. In support of this observation, prophylactic or therapeutic immunization of the mice with recombinant Pep1p could improve their survival when infected with a lethal dose of C. neoformans. Moreover, passive therapy with monoclonal anti-Pep1p antibodies also enhanced survival of the mice from C. neoformans infection. The associated antifungal mechanisms were mounting of a protective immune response and the development of fungal specific antibodies that decrease the fungal burden due to an increase in their phagocytosis and/or inhibit the fungal multiplication. Together, our study demonstrates (a) the mode of host-fungal interaction and the immune response developed thereby play a crucial role in developing resistance against C. neoformans; (b) Pep1p, an aspartic protease as well as an antigenic protein secreted by C. neoformans, can be exploited for vaccination (both prophylactic and therapeutic) or immunotherapy to improve the host defense during this fungal infection.
Collapse
Affiliation(s)
| | - Christine Laurent-Winter
- Institut Pasteur, Université Paris Cité, CNRS UAR 2024, Mass Spectrometry for Biology Unit, Proteomics platform, Paris, France
| | - Laurence Fiette
- Institut Pasteur, Human Histopathology and Animal Models Unit, Paris, France
| | - Guilhem Janbon
- Institut Pasteur, Université Paris Cité, CNRS, Molecular Mycology Unit, UMR 2000, Paris, France
| | - Vishukumar Aimanianda
- Institut Pasteur, Université Paris Cité, CNRS, Molecular Mycology Unit, UMR 2000, Paris, France
| | - Françoise Dromer
- Institut Pasteur, Université Paris Cité, CNRS, Molecular Mycology Unit, UMR 2000, Paris, France
| |
Collapse
|
4
|
Nenciarini S, Rivero D, Ciccione A, Amoriello R, Cerasuolo B, Pallecchi M, Bartolucci GL, Ballerini C, Cavalieri D. Impact of cooperative or competitive dynamics between the yeast Saccharomyces cerevisiae and lactobacilli on the immune response of the host. Front Immunol 2024; 15:1399842. [PMID: 39450162 PMCID: PMC11499123 DOI: 10.3389/fimmu.2024.1399842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 09/25/2024] [Indexed: 10/26/2024] Open
Abstract
Fungi and bacteria can be found coexisting in a wide variety of environments. The combination of their physical and molecular interactions can result in a broad range of outcomes for each partner, from competition to cooperative relationships. Most of these interactions can also be found in the human gastrointestinal tract. The gut microbiota is essential for humans, helping the assimilation of food components as well as the prevention of pathogen invasions through host immune system modulation and the production of beneficial metabolites such as short-chain fatty acids (SCFAs). Several factors, including changes in diet habits due to the progressive Westernization of the lifestyle, are linked to the onset of dysbiosis statuses that impair the correct balance of the gut environment. It is therefore crucial to explore the interactions between commensal and diet-derived microorganisms and their influence on host health. Investigating these interactions through co-cultures between human- and fermented food-derived lactobacilli and yeasts led us to understand how the strains' growth yield and their metabolic products rely on the nature and concentration of the species involved, producing either cooperative or competitive dynamics. Moreover, single cultures of yeasts and lactobacilli proved to be ideal candidates for developing immune-enhancing products, given their ability to induce trained immunity in blood-derived human monocytes in vitro. Conversely, co-cultures as well as mixtures of yeasts and lactobacilli have been shown to induce an anti-inflammatory response on the same immune cells in terms of cytokine profiles and activation surface markers, opening new possibilities in the design of probiotic and dietary therapies.
Collapse
Affiliation(s)
| | - Damariz Rivero
- Department of Biology, University of Florence, Firenze, Italy
| | | | - Roberta Amoriello
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | - Marco Pallecchi
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Gian Luca Bartolucci
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Clara Ballerini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Duccio Cavalieri
- Department of Biology, University of Florence, Firenze, Italy
- Interuniversity Consortium for Biotechnologies, Trieste, Italy
| |
Collapse
|
5
|
Swenson KA, Min K, Konopka JB. Candida albicans pathways that protect against organic peroxides and lipid peroxidation. PLoS Genet 2024; 20:e1011455. [PMID: 39432552 DOI: 10.1371/journal.pgen.1011455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/31/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024] Open
Abstract
Human fungal pathogens must survive diverse reactive oxygen species (ROS) produced by host immune cells that can oxidize a range of cellular molecules including proteins, lipids, and DNA. Formation of lipid radicals can be especially damaging, as it leads to a chain reaction of lipid peroxidation that causes widespread damage to the plasma membrane. Most previous studies on antioxidant pathways in fungal pathogens have been conducted with hydrogen peroxide, so the pathways used to combat organic peroxides and lipid peroxidation are not well understood. The most well-known peroxidase in Candida albicans, catalase, can only act on hydrogen peroxide. We therefore characterized a family of four glutathione peroxidases (GPxs) that were predicted to play an important role in reducing organic peroxides. One of the GPxs, Gpx3 is also known to activate the Cap1 transcription factor that plays the major role in inducing antioxidant genes in response to ROS. Surprisingly, we found that the only measurable role of the GPxs is activation of Cap1 and did not find a significant role for GPxs in the direct detoxification of peroxides. Furthermore, a CAP1 deletion mutant strain was highly sensitive to organic peroxides and oxidized lipids, indicating an important role for antioxidant genes upregulated by Cap1 in protecting cells from organic peroxides. We identified GLR1 (Glutathione reductase), a gene upregulated by Cap1, as important for protecting cells from oxidized lipids, implicating glutathione utilizing enzymes in the protection against lipid peroxidation. Furthermore, an RNA-sequencing study in C. albicans showed upregulation of a diverse set of antioxidant genes and protein damage pathways in response to organic peroxides. Overall, our results identify novel mechanisms by which C. albicans responds to oxidative stress resistance which open new avenues for understanding how fungal pathogens resist ROS in the host.
Collapse
Affiliation(s)
- Kara A Swenson
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, United States of America
| | - Kyunghun Min
- Department of Plant Science, Gangneung-Wonju National University, Gangneung, Republic of Korea
| | - James B Konopka
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, United States of America
| |
Collapse
|
6
|
Antunes D, Domingues R, Cruz-Almeida M, Rodrigues L, Borges O, Carvalho A, Casadevall A, Fernandes C, Gonçalves T. Cell wall nanoparticles from hyphae of Alternaria infectoria grown with caspofungin, nikkomycin, or pyroquilon trigger different activation profiles in macrophages. Microbiol Spectr 2024:e0064524. [PMID: 39329485 DOI: 10.1128/spectrum.00645-24] [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: 03/11/2024] [Accepted: 08/02/2024] [Indexed: 09/28/2024] Open
Abstract
Alternaria infectoria causes opportunistic human infections and is a source of allergens leading to respiratory allergies. In this work, we prepared cell wall nanoparticles (CWNPs) as a novel approach to study macrophage immunomodulation by fungal hyphal cell walls. A. infectoria was grown in the presence of caspofungin, an inhibitor of β(1,3)-glucan synthesis; nikkomycin Z, an inhibitor of chitin synthases; and pyroquilon, an inhibitor of dihydroxynaphthalene (DHN)-melanin synthesis. Distinct CWNPs were obtained from these cultures, referred to as casCWNPs, nkCWNPs, and pyrCWNPs, respectively. CWNPs are round-shaped particles with a diameter of 70-200 nm diameter particles that when added to macrophages are taken up by membrane ruffling. CWNPs with no DHN-melanin and more glucan (pyrCWNPs) caused early macrophage activation and lowest viability, with the cells exhibiting ultrastructural modifications such as higher vacuolization and formation of autophagy-like structures. CasCWNPs promoted the highest tumor necrosis factor alpha (TNF-α) and interleukin 1 beta (IL-1β) increase, also resulting in the release of partially degraded chitin, an aspect never observed in macrophage-like cells and fungi. After 6 h of interaction with CWNPs, only half were viable, except with control CWNPs. Overall, this work indicates that compounds that modify the fungal cell wall led to CWNPs with new properties that may have implications for the effects of drugs during antifungal therapy. CWNPs provide a new tool to study the interaction of hyphal fungal cell wall components with phagocytic cells and enable to show how the modification of cell wall components in A. infectoria can modulate the response by macrophages.IMPORTANCEAlternaria species are ubiquitous environmental fungi to which the human host can continuously be exposed, through the inhalation of fungal spores but also of fragments of hyphae, from desegregated mycelia. These fungi are involved in hypersensitization and severe respiratory allergies, such as asthma, and can cause opportunistic infections in immunodepressed human host leading to severe disease. The first fungal structures to interact with the host cells are the cell wall components, and their modulation leads to differential immune responses. Here, we show that fungal cells grown with cell wall inhibitors led to cell wall nanoparticles with new properties in their interaction with macrophages. With this strategy, we overcame the limitation of in vitro assays interacting with filamentous fungi and showed that the absence of DNH-melanin leads to higher virulence, while caspofungin leads to cells walls that trigger higher hydrolysis of chitin and higher production of cytokines.
Collapse
Affiliation(s)
- Daniela Antunes
- Univ Coimbra, CNC-UC-Center for Neuroscience and Cell Biology of the University of Coimbra, Coimbra, Portugal
- CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Rita Domingues
- Univ Coimbra, CNC-UC-Center for Neuroscience and Cell Biology of the University of Coimbra, Coimbra, Portugal
- CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Mariana Cruz-Almeida
- Univ Coimbra, CNC-UC-Center for Neuroscience and Cell Biology of the University of Coimbra, Coimbra, Portugal
- CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Lisa Rodrigues
- Univ Coimbra, CNC-UC-Center for Neuroscience and Cell Biology of the University of Coimbra, Coimbra, Portugal
- CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Olga Borges
- Univ Coimbra, CNC-UC-Center for Neuroscience and Cell Biology of the University of Coimbra, Coimbra, Portugal
- CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Univ Coimbra, FFUC-Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Chantal Fernandes
- Univ Coimbra, CNC-UC-Center for Neuroscience and Cell Biology of the University of Coimbra, Coimbra, Portugal
- CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Teresa Gonçalves
- Univ Coimbra, CNC-UC-Center for Neuroscience and Cell Biology of the University of Coimbra, Coimbra, Portugal
- CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Univ Coimbra, FMUC-Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
7
|
Rosati D, Pradhan A, van Heck JIP, Helder L, Jaeger M, Gow NAR, Joosten LAB, Williams DL, Brown AJP, Bruno M, Netea MG. Candida albicans N-Linked Mannans Potentiate the Induction of Trained Immunity via Dectin-2. J Infect Dis 2024; 230:768-777. [PMID: 38446996 PMCID: PMC11420807 DOI: 10.1093/infdis/jiae112] [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: 07/28/2023] [Revised: 01/23/2024] [Accepted: 02/28/2024] [Indexed: 03/08/2024] Open
Abstract
The interaction between the Candida albicans cell wall and pattern recognition receptors is crucial for the initiation of host immune responses, which, ultimately, contribute to the clearance of this pathogenic fungus. In the present study, we investigate the ability of C. albicans mannans to modulate immune response and induce innate immune memory (also termed trained immunity). Using mutants of C. albicans that are defective in or lack mannosyl residues, we show that alterations in the mannosylation of the C. albicans cell wall affect the innate cytokine response and strongly reduce the secretion of T-cell-derived cytokines. Subsequently, we demonstrate that the branching of N-linked mannan, but not O-linked mannan, is essential to potentiate the induction of trained immunity, a process mediated by dectin 2. In conclusion, N-linked mannan is needed, in addition to β-glucans, for an effective induction of trained immunity by C. albicans.
Collapse
Affiliation(s)
- Diletta Rosati
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, the Netherlands
| | - Arnab Pradhan
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter EX4 4QD, United Kingdom
| | - Julia I P van Heck
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, the Netherlands
| | - Leonie Helder
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, the Netherlands
| | - Martin Jaeger
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, the Netherlands
| | - Neil A R Gow
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter EX4 4QD, United Kingdom
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, the Netherlands
- Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy Cluj-Napoca, Romania
| | - David L Williams
- Departments of Surgery, Biomedical Sciences and Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Alistair J P Brown
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter EX4 4QD, United Kingdom
| | - Mariolina Bruno
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, the Netherlands
- Department for Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| |
Collapse
|
8
|
Chatterjee P, Moss CT, Omar S, Dhillon E, Hernandez Borges CD, Tang AC, Stevens DA, Hsu JL. Allergic Bronchopulmonary Aspergillosis (ABPA) in the Era of Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) Modulators. J Fungi (Basel) 2024; 10:656. [PMID: 39330416 PMCID: PMC11433030 DOI: 10.3390/jof10090656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
Allergic bronchopulmonary aspergillosis (ABPA) is a hypersensitivity disease caused by Aspergillus fumigatus (Af), prevalent in persons with cystic fibrosis (CF) or asthma. In ABPA, Af proteases drive a T-helper cell-2 (Th2)-mediated allergic immune response leading to inflammation that contributes to permanent lung damage. Corticosteroids and antifungals are the mainstays of therapies for ABPA. However, their long-term use has negative sequelae. The treatment of patients with CF (pwCF) has been revolutionized by the efficacy of cystic fibrosis transmembrane conductance regulator (CFTR) modulator therapy. Pharmacological improvement in CFTR function with highly effective elexacaftor/tezacaftor/ivacaftor (ETI) provides unprecedented improvements in lung function and other clinical outcomes of pwCF. The mechanism behind the improvement in patient outcomes is a continued topic of investigation as our understanding of the role of CFTR function evolves. As ETI therapy gains traction in CF management, understanding its potential impact on ABPA, especially on the allergic immune response pathways and Af infection becomes increasingly crucial for optimizing patient outcomes. This literature review aims to examine the extent of these findings and expand our understanding of the already published research focusing on the intersection between ABPA therapeutic approaches in CF and the rapid impact of the evolving CFTR modulator landscape. While our literature search yielded limited reports specifically focusing on the role of CFTR modulator therapy on CF-ABPA, findings from epidemiologic and retrospective studies suggest the potential for CFTR modulator therapies to positively influence pulmonary outcomes by addressing the underlying pathophysiology of CF-ABPA, especially by decreasing inflammatory response and Af colonization. Thus, this review highlights the promising scope of CFTR modulator therapy in decreasing the overall prevalence and incidence of CF-ABPA.
Collapse
Affiliation(s)
- Paulami Chatterjee
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (P.C.); (S.O.); (E.D.)
| | - Carson Tyler Moss
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Sarah Omar
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (P.C.); (S.O.); (E.D.)
| | - Ekroop Dhillon
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (P.C.); (S.O.); (E.D.)
| | | | - Alan C. Tang
- Department of Medicine, Keck School of Medicine, Los Angeles, CA 90089, USA;
| | - David A. Stevens
- Division of Infectious Diseases and Geographic Medicine, Stanford University Medical School, Stanford, CA 94305, USA;
| | - Joe L. Hsu
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (P.C.); (S.O.); (E.D.)
| |
Collapse
|
9
|
Bihaniya H, Rudraprasad D, Joseph J. Pathobiology of Fungal Endophthalmitis: A Major Review. ACS Infect Dis 2024; 10:3126-3137. [PMID: 39267469 DOI: 10.1021/acsinfecdis.4c00442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
Fungal endophthalmitis is a chronic inflammatory condition of the eye's posterior segment that can lead to irreversible vision loss. While relatively rare in western countries, its incidence is notably higher in Asia, particularly India. The condition's prevalence is exacerbated by factors such as intravenous drug use, antibiotics, and ocular surgeries. Fungal endophthalmitis can be categorized as endogenous, arising from systemic infection, or exogenous, linked to external sources such as trauma or surgery. The fungal agents responsible vary by region, with Candida species common in the West and Aspergillus and Fusarium species more prevalent in India. Management typically involves vitrectomy and intravitreal antifungal drugs such as amphotericin B and voriconazole, though treatment is often complicated by multidrug resistance and culture-negative cases. Recent proteomic and transcriptomic analyses have highlighted the early and sustained activation of the host immune response during infection involving key inflammatory and oxidative stress-related proteins. Given the potential for excessive inflammation to cause retinal damage, targeted immunotherapies are crucial. Immunomodulation, which aims to balance the immune response, shows promise in preserving vision while effectively combating the infection. Key targets for immunomodulation include pro-inflammatory cytokines (TNF-α, IL-1β, IL-6, IL-17), chemokines (CCL2, CXCL8), Toll-like receptors (TLR2, TLR4), and the complement system. Additionally, modulating the activity of macrophages, neutrophils, regulatory T cells, and Th17 cells, as well as targeting inflammasomes, can help control inflammation. Biologic agents and small molecule inhibitors offer further avenues for precise immune response modulation. This review underscores the importance of a comprehensive understanding of host-pathogen interactions in the development of effective therapies for fungal endophthalmitis.
Collapse
Affiliation(s)
- Himanshi Bihaniya
- Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad, Telangana 500034, India
| | - Dhanwini Rudraprasad
- Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad, Telangana 500034, India
- Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Joveeta Joseph
- Jhaveri Microbiology Centre, Brien Holden Eye Research Centre, L. V. Prasad Eye Institute, Hyderabad, Telangana 500034, India
| |
Collapse
|
10
|
Zhang X, Hu D, Sun X, Gu Y, Zhou Y, Su C, Liu S, Zhang C, Lu G, Wu Q, Chen A. PHGDH/SYK: a hub integrating anti-fungal immunity and serine metabolism. Cell Death Differ 2024:10.1038/s41418-024-01374-7. [PMID: 39256519 DOI: 10.1038/s41418-024-01374-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/12/2024] Open
Abstract
Immune cells modify their metabolic pathways in response to fungal infections. Nevertheless, the biochemical underpinnings need to be better understood. This study reports that fungal infection drives a switch from glycolysis to the serine synthesis pathway (SSP) and one-carbon metabolism by inducing the interaction of spleen tyrosine kinase (SYK) and phosphoglycerate dehydrogenase (PHGDH). As a result, PHGDH promotes SYK phosphorylation, leading to the recruitment of SYK to C-type lectin receptors (CLRs). The CLR/SYK complex initiates signaling cascades that lead to transcription factor activation and pro-inflammatory cytokine production. SYK activates SSP and one-carbon metabolism by inducing PHGDH activity. Then, one-carbon metabolism supports S-adenosylmethionine and histone H3 lysine 36 trimethylation to drive the production of pro-inflammatory cytokines and chemokines. These findings reveal the crosstalk between amino acid metabolism, epigenetic modification, and CLR signaling during fungal infection.
Collapse
Affiliation(s)
- Xinyong Zhang
- Department of Neurology, The Second People's Hospital of Huai 'an, Huai 'an, 223001, China
| | - Dongdong Hu
- Department of Emergency, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, 210031, China
| | - Xiaoyan Sun
- School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, 316022, China
| | - Yichun Gu
- The Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Yong Zhou
- Department of Neurology, The Second People's Hospital of Huai 'an, Huai 'an, 223001, China
| | - Chuanxin Su
- The Key Laboratory of Targeted Intervention of Clinical Disease, University Hospital Essen, University of Duisburg-Essen, Essen, 45122, Germany
| | - Shi Liu
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Caiyan Zhang
- Department of Critical Care Medicine, Children's Hospital of Fudan University, Shanghai, China
| | - Guoping Lu
- Department of Critical Care Medicine, Children's Hospital of Fudan University, Shanghai, China
| | - Qiwen Wu
- Department of Laboratory Medicine, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, China.
| | - Aidong Chen
- The Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
11
|
Wahab A, Sanborn D, Vergidis P, Razonazole R, Yadav H, Pennington KM. Diagnosis and Prevention of Invasive Fungal Infections in the Immunocompromised Host. Chest 2024:S0012-3692(24)05140-7. [PMID: 39245320 DOI: 10.1016/j.chest.2024.08.046] [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: 07/11/2023] [Revised: 08/16/2024] [Accepted: 08/30/2024] [Indexed: 09/10/2024] Open
Abstract
TOPIC IMPORTANCE The prevalence of invasive fungal infections (IFIs) has risen in the past 3 decades, attributed to advancements in immune-modulatory therapies used in transplantation, rheumatology, and oncology. REVIEW FINDINGS Organisms that cause IFI evade the host's natural defenses or at opportunities of immunologic weakness. Infections occur from inhalation of potentially pathogenic organisms, translocation of commensal organisms, or reactivation of latent infection. Organisms that cause IFI in immunocompromised populations include Candida species, Cryptococcus species, environmental molds, and endemic fungi. Diagnosis of these infections is challenging due to slow organism growth and fastidious culture requirements. Moreover, fungal biomarkers tend to be nonspecific and can be negatively impacted by prophylactic antifungals. Antibody-based tests are not sensitive in immunocompromised hosts making antigen-based testing necessary. Risk reduction of IFI is guided by pathogen avoidance, removal or minimization of immune-suppressing factors, and pharmacologic prophylaxis in select hosts. SUMMARY Understanding the complex interplay between the immune system and opportunistic fungal pathogens plays a key role in early diagnosis and prevention.
Collapse
Affiliation(s)
- Abdul Wahab
- Department of Medicine, Mayo Clinic Health Systems, Mankato, MN
| | - David Sanborn
- Divisions of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN
| | - Paschalis Vergidis
- Infectious Disease, Mayo Clinic, Rochester, MN; William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN
| | - Raymund Razonazole
- Infectious Disease, Mayo Clinic, Rochester, MN; William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN
| | - Hemang Yadav
- Divisions of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN; William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN
| | - Kelly M Pennington
- Divisions of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN; William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN.
| |
Collapse
|
12
|
Rinker DC, Sauters TJC, Steffen K, Gumilang A, Raja HA, Rangel-Grimaldo M, Pinzan CF, de Castro PA, Dos Reis TF, Delbaje E, Houbraken J, Goldman GH, Oberlies NH, Rokas A. Strain heterogeneity in a non-pathogenic Aspergillus fungus highlights factors associated with virulence. Commun Biol 2024; 7:1082. [PMID: 39232082 PMCID: PMC11374809 DOI: 10.1038/s42003-024-06756-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024] Open
Abstract
Fungal pathogens exhibit extensive strain heterogeneity, including variation in virulence. Whether closely related non-pathogenic species also exhibit strain heterogeneity remains unknown. Here, we comprehensively characterized the pathogenic potentials (i.e., the ability to cause morbidity and mortality) of 16 diverse strains of Aspergillus fischeri, a non-pathogenic close relative of the major pathogen Aspergillus fumigatus. In vitro immune response assays and in vivo virulence assays using a mouse model of pulmonary aspergillosis showed that A. fischeri strains varied widely in their pathogenic potential. Furthermore, pangenome analyses suggest that A. fischeri genomic and phenotypic diversity is even greater. Genomic, transcriptomic, and metabolic profiling identified several pathways and secondary metabolites associated with variation in virulence. Notably, strain virulence was associated with the simultaneous presence of the secondary metabolites hexadehydroastechrome and gliotoxin. We submit that examining the pathogenic potentials of non-pathogenic close relatives is key for understanding the origins of fungal pathogenicity.
Collapse
Affiliation(s)
- David C Rinker
- Department of Biological Sciences and Evolutionary Studies Initiative, Vanderbilt University, Nashville, TN, USA
| | - Thomas J C Sauters
- Department of Biological Sciences and Evolutionary Studies Initiative, Vanderbilt University, Nashville, TN, USA
| | - Karin Steffen
- Department of Biological Sciences and Evolutionary Studies Initiative, Vanderbilt University, Nashville, TN, USA
| | - Adiyantara Gumilang
- Department of Biological Sciences and Evolutionary Studies Initiative, Vanderbilt University, Nashville, TN, USA
| | - Huzefa A Raja
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC, USA
| | - Manuel Rangel-Grimaldo
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC, USA
| | - Camila Figueiredo Pinzan
- Faculdade de Ciencias Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Patrícia Alves de Castro
- Faculdade de Ciencias Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Thaila Fernanda Dos Reis
- Faculdade de Ciencias Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Endrews Delbaje
- Faculdade de Ciencias Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Jos Houbraken
- Food and Indoor Mycology, Westerdijk Fungal Biodiversity Institute, Utrecht, The Netherlands
| | - Gustavo H Goldman
- Faculdade de Ciencias Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil.
| | - Nicholas H Oberlies
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC, USA.
| | - Antonis Rokas
- Department of Biological Sciences and Evolutionary Studies Initiative, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
13
|
Kwait R, Pinsky ML, Gignoux‐Wolfsohn S, Eskew EA, Kerwin K, Maslo B. Impact of putatively beneficial genomic loci on gene expression in little brown bats ( Myotis lucifugus, Le Conte, 1831) affected by white-nose syndrome. Evol Appl 2024; 17:e13748. [PMID: 39310794 PMCID: PMC11413065 DOI: 10.1111/eva.13748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 06/06/2024] [Accepted: 06/19/2024] [Indexed: 09/25/2024] Open
Abstract
Genome-wide scans for selection have become a popular tool for investigating evolutionary responses in wildlife to emerging diseases. However, genome scans are susceptible to false positives and do little to demonstrate specific mechanisms by which loci impact survival. Linking putatively resistant genotypes to observable phenotypes increases confidence in genome scan results and provides evidence of survival mechanisms that can guide conservation and management efforts. Here we used an expression quantitative trait loci (eQTL) analysis to uncover relationships between gene expression and alleles associated with the survival of little brown bats (Myotis lucifugus) despite infection with the causative agent of white-nose syndrome. We found that 25 of the 63 single-nucleotide polymorphisms (SNPs) associated with survival were related to gene expression in wing tissue. The differentially expressed genes have functional annotations associated with the innate immune system, metabolism, circadian rhythms, and the cellular response to stress. In addition, we observed differential expression of multiple genes with survival implications related to loci in linkage disequilibrium with focal SNPs. Together, these findings support the selective function of these loci and suggest that part of the mechanism driving survival may be the alteration of immune and other responses in epithelial tissue.
Collapse
Affiliation(s)
- Robert Kwait
- Department of Ecology, Evolution and Natural ResourcesRutgers, The State University of New JerseyNew BrunswickNew JerseyUSA
| | - Malin L. Pinsky
- Department of Ecology, Evolution and Natural ResourcesRutgers, The State University of New JerseyNew BrunswickNew JerseyUSA
- Department of Ecology and Evolutionary BiologyUniversity of California Santa CruzSanta CruzCaliforniaUSA
| | | | - Evan A. Eskew
- Institute for Interdisciplinary Data SciencesUniversity of IdahoMoscowIdahoUSA
| | - Kathleen Kerwin
- Department of Ecology, Evolution and Natural ResourcesRutgers, The State University of New JerseyNew BrunswickNew JerseyUSA
| | - Brooke Maslo
- Department of Ecology, Evolution and Natural ResourcesRutgers, The State University of New JerseyNew BrunswickNew JerseyUSA
| |
Collapse
|
14
|
Jia LJ, González K, Orasch T, Schmidt F, Brakhage AA. Manipulation of host phagocytosis by fungal pathogens and therapeutic opportunities. Nat Microbiol 2024; 9:2216-2231. [PMID: 39187614 DOI: 10.1038/s41564-024-01780-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 07/09/2024] [Indexed: 08/28/2024]
Abstract
An important host defence mechanism against pathogens is intracellular killing, which is achieved through phagocytosis, a cellular process for engulfing and neutralizing extracellular particles. Phagocytosis results in the formation of matured phagolysosomes, which are specialized compartments that provide a hostile environment and are considered the end point of the degradative pathway. However, all fungal pathogens studied to date have developed strategies to manipulate phagosomal function directly and also indirectly by redirecting phagosomes from the degradative pathway to a non-degradative pathway with the expulsion and even transfer of pathogens between cells. Here, using the major human fungal pathogens Aspergillus fumigatus, Candida albicans, Cryptococcus neoformans and Histoplasma capsulatum as examples, we discuss the processes involved in host phagosome-fungal pathogen interactions, with a focus on fungal evasion strategies. We also discuss recent approaches to targeting intraphagosomal pathogens, including the redirection of phagosomes towards degradative pathways for fungal pathogen eradication.
Collapse
Affiliation(s)
- Lei-Jie Jia
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (Leibniz-HKI), Jena, Germany.
- Junior Research Group Phagosome Biology and Engineering, Leibniz-HKI, Jena, Germany.
| | - Katherine González
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (Leibniz-HKI), Jena, Germany
| | - Thomas Orasch
- Transfer Group Anti-infectives, Leibniz-HKI, Jena, Germany
| | - Franziska Schmidt
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (Leibniz-HKI), Jena, Germany
| | - Axel A Brakhage
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (Leibniz-HKI), Jena, Germany.
- Department of Microbiology and Molecular Biology, Institute of Microbiology, Friedrich Schiller University, Jena, Germany.
| |
Collapse
|
15
|
Omer I, Khalil I, Abdalmumin A, Molefe PF, Sabeel S, Farh IZA, Mohamed HA, Elsharif HA, Mohamed ALAH, Awad‐Elkareem MA, Salih M. Design of an epitope-based peptide vaccine against Cryptococcus neoformans. FEBS Open Bio 2024; 14:1471-1489. [PMID: 39020466 PMCID: PMC11492362 DOI: 10.1002/2211-5463.13858] [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: 01/20/2023] [Revised: 04/11/2024] [Accepted: 06/21/2024] [Indexed: 07/19/2024] Open
Abstract
Cryptococcus neoformans is the highest-ranked fungal pathogen in the Fungal Priority Pathogens List (FPPL) released by the World Health Organization (WHO). In this study, through in silico simulations, a multi-epitope vaccine against Cryptococcus neoformans was developed using the mannoprotein antigen (MP88) as a vaccine candidate. Following the retrieval of the MP88 protein sequences, these were used to predict antigenic B-cell and T-cell epitopes via the bepipred tool and the artificial neural network, respectively. Conserved B-cell epitopes AYSTPA, AYSTPAS, PASSNCK, and DSAYPP were identified as the most promising B-cell epitopes. While YMAADQFCL, VSYEEWMNY, and FQQRYTGTF were identified as the best candidates for CD8+ T-cell epitopes; and YARLLSLNA, ISYGTAMAV, and INQTSYARL were identified as the most promising CD4+ T-cell epitopes. The vaccine construct was modeled along with adjuvant and peptide linkers and the expasy protparam tool was used to predict the physiochemical properties. According to this, the construct vaccine was predicted to be antigenic, nontoxic, nonallergenic, soluble, stable, hydrophilic, and thermostable. Furthermore, the three-dimensional structure was also used in docking analyses with Toll-like receptor (TLR4). Finally, the cDNA of vaccine was successfully cloned into the E. coli pET-28a (+) expression vector. The results presented here could contribute towards the design of an effective vaccine against Cryptococcus neoformans.
Collapse
Affiliation(s)
- Ibtihal Omer
- Department of Therapeutic Drug Monitoring LaboratoryNational Center for Kidney Diseases and SurgeryKhartoumSudan
| | - Isra Khalil
- Department of Microbiology, Faculty of Medical Laboratory ScienceSudan University of Science and TechnologyKhartoumSudan
| | - Ahmed Abdalmumin
- Biomedical Research InstituteSudan National UniversityKhartoumSudan
| | - Philisiwe Fortunate Molefe
- Hair and Skin Research Laboratory, Department of Medicine, Division Dermatology, Groote Schuur HospitalUniversity of Cape TownCape TownSouth Africa
| | - Solima Sabeel
- Department of Pathology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine (IDM)University of Cape TownSouth Africa
| | | | - Hanaa Abdalla Mohamed
- Department of Microbiology, Faculty of Medical Laboratory ScienceSudan University of Science and TechnologyKhartoumSudan
| | - Hajr Abdallha Elsharif
- General Administration of Quarantine and Animal HealthRegional Training InstituteKhartoumSudan
| | | | | | - Mohamed Salih
- Department of BiotechnologyAfrica City of TechnologyKhartoumSudan
| |
Collapse
|
16
|
da Fonseca DM, Rodrigues L, Sousa-Baptista J, Marcos-Tejedor F, Mota M, Cunha RA, Fernandes C, Gonçalves T. Caffeine Protects Keratinocytes from Trichophyton mentagrophytes Infection and Behaves as an Antidermatophytic Agent. Int J Mol Sci 2024; 25:8303. [PMID: 39125871 PMCID: PMC11311904 DOI: 10.3390/ijms25158303] [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: 06/22/2024] [Revised: 07/21/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Caffeine affords several beneficial effects on human health, acting as an antioxidant, anti-inflammatory agent, and analgesic. Caffeine is widely used in cosmetics, but its antimicrobial activity has been scarcely explored, namely against skin infection agents. Dermatophytes are the most common fungal agents of human infection, mainly of skin infections. This work describes the in vitro effect of caffeine during keratinocyte infection by Trichophyton mentagrophytes, one of the most common dermatophytes. The results show that caffeine was endowed with antidermatophytic activity with a MIC, determined following the EUCAST standards, of 8 mM. Caffeine triggered a modification of the levels of two major components of the fungal cell wall, β-(1,3)-glucan and chitin. Caffeine also disturbed the ultrastructure of the fungal cells, particularly the cell wall surface and mitochondria, and autophagic-like structures were observed. During dermatophyte-human keratinocyte interactions, caffeine prevented the loss of viability of keratinocytes and delayed spore germination. Overall, this indicates that caffeine can act as a therapeutic and prophylactic agent for dermatophytosis.
Collapse
Affiliation(s)
- Diogo M. da Fonseca
- FMUC—Faculty of Medicine, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal; (D.M.d.F.); (J.S.-B.); (M.M.); (R.A.C.)
- CNC-UC—Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; (L.R.); (C.F.)
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Lisa Rodrigues
- CNC-UC—Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; (L.R.); (C.F.)
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - José Sousa-Baptista
- FMUC—Faculty of Medicine, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal; (D.M.d.F.); (J.S.-B.); (M.M.); (R.A.C.)
- CNC-UC—Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; (L.R.); (C.F.)
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Félix Marcos-Tejedor
- Department of Medical Sciences, Faculty of Health Sciences, University of Castilla-La Mancha, 45600 Talavera de la Reina, Toledo, Spain;
| | - Marta Mota
- FMUC—Faculty of Medicine, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal; (D.M.d.F.); (J.S.-B.); (M.M.); (R.A.C.)
- CNC-UC—Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; (L.R.); (C.F.)
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Rodrigo A. Cunha
- FMUC—Faculty of Medicine, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal; (D.M.d.F.); (J.S.-B.); (M.M.); (R.A.C.)
- CNC-UC—Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; (L.R.); (C.F.)
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Chantal Fernandes
- CNC-UC—Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; (L.R.); (C.F.)
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Teresa Gonçalves
- FMUC—Faculty of Medicine, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal; (D.M.d.F.); (J.S.-B.); (M.M.); (R.A.C.)
- CNC-UC—Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; (L.R.); (C.F.)
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
17
|
Oliveira MDA, de Almeida SR, Martins JO. Novel Insights into Sporotrichosis and Diabetes. J Fungi (Basel) 2024; 10:527. [PMID: 39194853 DOI: 10.3390/jof10080527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
Sporotrichosis is a type of zoonotic subcutaneous mycosis caused by different species of dimorphic fungus of the genus Sporothrix, and it is the most common form of subcutaneous mycosis in Latin America. Sporotrichosis is generally restricted to cutaneous and lymphatic tissue (i.e., localized forms), and involvement in the viscera (i.e., disseminated or disseminated cutaneous form) is uncommon, especially in the central nervous system. However, immunosuppression in individuals with diabetes mellitus can lead to the disseminated form of the disease due to a failure to eliminate the pathogen and poor infection treatment outcomes. Possible correlations between patients with diabetes and their greater susceptibility to disseminated cases of sporotrichosis include a decreased cytokine response after stimulation, increased oxidative stress, decreased chemotaxis, phagocytic activity, adhesion and rolling of neutrophils and monocytes/macrophages, and increased macrophage/monocyte and polymorphonuclear cell apoptosis. Therefore, this review highlights novel insights into diabetes and sporotrichosis by investigating how chronic inflammation affects and aggravates the infection, the possible causes of the greater susceptibility of Sporothrix sp. to hematogenous dissemination in immunocompromised patients, and the main alterations that this dissemination can cause.
Collapse
Affiliation(s)
- Mariana de Araujo Oliveira
- Laboratory of Immunoendocrinology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil
| | - Sandro Rogério de Almeida
- Laboratory of Mycology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil
| | - Joilson O Martins
- Laboratory of Immunoendocrinology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil
| |
Collapse
|
18
|
Bettelli F, Vallerini D, Lagreca I, Barozzi P, Riva G, Nasillo V, Paolini A, D'Amico R, Forghieri F, Morselli M, Pioli V, Gilioli A, Giusti D, Messerotti A, Bresciani P, Cuoghi A, Colaci E, Marasca R, Pagano L, Candoni A, Maertens J, Viale P, Mussini C, Manfredini R, Tagliafico E, Sarti M, Trenti T, Lewis R, Comoli P, Eccher A, Luppi M, Potenza L. Identification and validation of diagnostic cut-offs of the ELISpot assay for the diagnosis of invasive aspergillosis in high-risk patients. PLoS One 2024; 19:e0306728. [PMID: 38980880 PMCID: PMC11233002 DOI: 10.1371/journal.pone.0306728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 06/21/2024] [Indexed: 07/11/2024] Open
Abstract
OBJECTIVE We investigated the performance of enzyme linked immunospot (ELISpot) assay for the diagnosis of invasive aspergillosis (IA) in high-risk patients with hematologic malignancies. METHODS We prospectively enrolled two cohorts of patients undergoing intensive myelosuppressive or immunosuppressive treatments at high risk for IA. ELISpot was performed to detect Aspergillus-specific T cells producing Interleukin-10. RESULTS In the discovery cohort, a derived cut-off of 40 spot forming cells (SFCs)/106 PBMCs has shown to correctly classify IA cases with a sensitivity and specificity of 89.5% and 88.6%, respectively. This cut-off is lowered to 25 SFC when considering the subset of possible IA patients, with sensitivity and specificity of 76% and 93%, respectively. The application of the 40 SFCs cut-off to the validation cohort resulted in a positivity rate of 83.3% in proven/probable cases and a negativity rate of 92.5% in possible/non-IA cases. Adopting the 25 SCFs cut-off, the assay resulted positive in 83.3% of proven/probable cases while it resulted negative in 66.7% of possible/non-IA cases. CONCLUSIONS ELISpot shows promises in the diagnosis of IA and the possibility to use two distinct cut-offs with similar diagnostic performances according to patients' different pre-test probability of infection can widen its use in patients at risk.
Collapse
Affiliation(s)
- Francesca Bettelli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, Modena, Italy
| | - Daniela Vallerini
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, Modena, Italy
| | - Ivana Lagreca
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, Modena, Italy
| | - Patrizia Barozzi
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, Modena, Italy
| | - Giovanni Riva
- Diagnostic Hematology and Clinical Genomics, Department of Laboratory Medicine and Pathology, AUSL/AOU Modena, Modena, Italy
| | - Vincenzo Nasillo
- Diagnostic Hematology and Clinical Genomics, Department of Laboratory Medicine and Pathology, AUSL/AOU Modena, Modena, Italy
| | - Ambra Paolini
- Diagnostic Hematology and Clinical Genomics, Department of Laboratory Medicine and Pathology, AUSL/AOU Modena, Modena, Italy
| | - Roberto D'Amico
- Statistic Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, Modena, Italy
| | - Fabio Forghieri
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, Modena, Italy
| | - Monica Morselli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, Modena, Italy
| | - Valeria Pioli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, Modena, Italy
| | - Andrea Gilioli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, Modena, Italy
| | - Davide Giusti
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, Modena, Italy
| | - Andrea Messerotti
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, Modena, Italy
| | - Paola Bresciani
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, Modena, Italy
| | - Angela Cuoghi
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, Modena, Italy
| | - Elisabetta Colaci
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, Modena, Italy
| | - Roberto Marasca
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, Modena, Italy
| | - Livio Pagano
- Hematology Division, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli - IRCCS and Università Cattolica del Sacro Cuore, Rome, Italy
| | - Anna Candoni
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, Modena, Italy
| | - Johan Maertens
- Department of Hematology, University Hospital UZ Leuven, Leuven, Belgium
| | - Pierluigi Viale
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, University of Bologna, IRCCS-AOU Policlinico Santorsola, Bologna, Italy
| | - Cristina Mussini
- Clinic of Infectious Diseases, Dipartimento Chirurgico, Medico, Odontoiatrico e di Scienze Morfologiche con interesse Trapiantologico, Oncologico e di Medicina Rigenerativa, University of Modena and Reggio Emilia, AOU Modena, Modena, Italy
| | - Rossella Manfredini
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Enrico Tagliafico
- Diagnostic Hematology and Clinical Genomics, Department of Laboratory Medicine and Pathology, AUSL/AOU Modena, Modena, Italy
| | - Mario Sarti
- Diagnostic Hematology and Clinical Genomics, Department of Laboratory Medicine and Pathology, AUSL/AOU Modena, Modena, Italy
| | - Tommaso Trenti
- Diagnostic Hematology and Clinical Genomics, Department of Laboratory Medicine and Pathology, AUSL/AOU Modena, Modena, Italy
| | - Russell Lewis
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Patrizia Comoli
- Pediatric Hematology/Oncology Unit and Cell Factory, Foundation IRCCS Policlinico San Matteo, Pavia, Italy
| | - Albino Eccher
- Section of Pathology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, Modena, Italy
| | - Mario Luppi
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, Modena, Italy
| | - Leonardo Potenza
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, Modena, Italy
| |
Collapse
|
19
|
Kaminski VL, Borges BM, Santos BV, Preite NW, Calich VLG, Loures FV. MDSCs use a complex molecular network to suppress T-cell immunity in a pulmonary model of fungal infection. Front Cell Infect Microbiol 2024; 14:1392744. [PMID: 39035356 PMCID: PMC11257977 DOI: 10.3389/fcimb.2024.1392744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/18/2024] [Indexed: 07/23/2024] Open
Abstract
Background Paracoccidioidomycosis (PCM) is a systemic endemic fungal disease prevalent in Latin America. Previous studies revealed that host immunity against PCM is tightly regulated by several suppressive mechanisms mediated by tolerogenic plasmacytoid dendritic cells, the enzyme 2,3 indoleamine dioxygenase (IDO-1), regulatory T-cells (Tregs), and through the recruitment and activation of myeloid-derived suppressor cells (MDSCs). We have recently shown that Dectin-1, TLR2, and TLR4 signaling influence the IDO-1-mediated suppression caused by MDSCs. However, the contribution of these receptors in the production of important immunosuppressive molecules used by MDSCs has not yet been explored in pulmonary PCM. Methods We evaluated the expression of PD-L1, IL-10, as well as nitrotyrosine by MDSCs after anti-Dectin-1, anti-TLR2, and anti-TLR4 antibody treatment followed by P. brasiliensis yeasts challenge in vitro. We also investigated the influence of PD-L1, IL-10, and nitrotyrosine in the suppressive activity of lung-infiltrating MDSCs of C57BL/6-WT, Dectin-1KO, TLR2KO, and TLR4KO mice after in vivo fungal infection. The suppressive activity of MDSCs was evaluated in cocultures of isolated MDSCs with activated T-cells. Results A reduced expression of IL-10 and nitrotyrosine was observed after in vitro anti-Dectin-1 treatment of MDSCs challenged with fungal cells. This finding was further confirmed in vitro and in vivo by using Dectin-1KO mice. Furthermore, MDSCs derived from Dectin-1KO mice showed a significantly reduced immunosuppressive activity on the proliferation of CD4+ and CD8+ T lymphocytes. Blocking of TLR2 and TLR4 by mAbs and using MDSCs from TLR2KO and TLR4KO mice also reduced the production of suppressive molecules induced by fungal challenge. In vitro, MDSCs from TLR4KO mice presented a reduced suppressive capacity over the proliferation of CD4+ T-cells. Conclusion We showed that the pathogen recognition receptors (PRRs) Dectin-1, TLR2, and TLR4 contribute to the suppressive activity of MDSCs by inducing the expression of several immunosuppressive molecules such as PD-L1, IL-10, and nitrotyrosine. This is the first demonstration of a complex network of PRRs signaling in the induction of several suppressive molecules by MDSCs and its contribution to the immunosuppressive mechanisms that control immunity and severity of pulmonary PCM.
Collapse
MESH Headings
- Animals
- Mice
- Interleukin-10/metabolism
- Toll-Like Receptor 2/metabolism
- Toll-Like Receptor 2/genetics
- Toll-Like Receptor 2/immunology
- Myeloid-Derived Suppressor Cells/immunology
- Myeloid-Derived Suppressor Cells/metabolism
- Toll-Like Receptor 4/metabolism
- Toll-Like Receptor 4/genetics
- Toll-Like Receptor 4/immunology
- Lectins, C-Type/metabolism
- Lectins, C-Type/genetics
- Disease Models, Animal
- B7-H1 Antigen/metabolism
- B7-H1 Antigen/genetics
- Mice, Inbred C57BL
- Paracoccidioidomycosis/immunology
- Paracoccidioides/immunology
- Tyrosine/analogs & derivatives
- Tyrosine/metabolism
- T-Lymphocytes, Regulatory/immunology
- Lung/immunology
- Lung/microbiology
- Signal Transduction
- Male
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Mice, Knockout
Collapse
Affiliation(s)
- Valéria Lima Kaminski
- Institute of Science and Technology, Federal University of São Paulo – UNIFESP, São Paulo, Brazil
| | - Bruno Montanari Borges
- Institute of Science and Technology, Federal University of São Paulo – UNIFESP, São Paulo, Brazil
| | - Bianca Vieira Santos
- Institute of Science and Technology, Federal University of São Paulo – UNIFESP, São Paulo, Brazil
| | - Nycolas Willian Preite
- Institute of Science and Technology, Federal University of São Paulo – UNIFESP, São Paulo, Brazil
| | - Vera Lucia Garcia Calich
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo – USP, São Paulo, Brazil
| | - Flávio Vieira Loures
- Institute of Science and Technology, Federal University of São Paulo – UNIFESP, São Paulo, Brazil
| |
Collapse
|
20
|
Pathakumari B, Liu W, Wang Q, Kong X, Liang G, Chokkakula S, Pathakamuri V, Nunna V. Comparative Evaluation of Candida Species-Specific T-Cell Immune Response in Human Peripheral Blood Mononuclear Cells. Biomedicines 2024; 12:1487. [PMID: 39062060 PMCID: PMC11274682 DOI: 10.3390/biomedicines12071487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/19/2024] [Accepted: 05/28/2024] [Indexed: 07/28/2024] Open
Abstract
Non-albicans Candida (NAC) species are increasingly recognized as significant contributors to candidemia infections; however, relatively less is known about the immune responses induced by these species. In this study, we compared the cytokine production ability of human peripheral blood mononuclear cells (PBMCs) upon stimulation with different Candida species (Candida spp.). We measured secreted cytokines using ELISA and checked the functional profiles of T-cell responses using multicolor flow cytometry. Although there was a differential expression of cytokines against Candida spp., significant difference were observed in the levels of IFN-γ, TNF-α, IL-10, IL-12p40, and IL-23 (p < 0.05) between Candida spp. A significant difference was observed between C. albicans and C. glabrata (p = 0.026) in the levels of TNF-α. C. glabrata showed significant differences compared to C. albicans, C. parapsilosis, and C. krusei in the levels of IL-10 (p values of 0.02, 0.04, and 0.01, respectively). Despite the percentages of CD4+ and CD8+ expressing Th1, Th2, and Th17 cytokines being higher in stimulated PBMCs, none of the Candida spp. showed significant differences. The levels of secreted IL-17A and IL-23 were consistently lower in Candida spp. regardless of the stimulus used. Here, we showed the differential regulation of Th1, Th2, and Th17 during Candida spp. stimulation of the immune system ex vivo. Additionally, our findings suggest that C. albicans elicits an IFN-γ response, whereas C. glabrata promotes IL-10 cellular responses, but this warrants additional studies to conclude this association. This investigation holds the potential to advance our comprehension of the distinct immune responses induced by Candida spp., with probable implications in designing antifungal immunotherapeutics.
Collapse
Affiliation(s)
- Balaji Pathakumari
- Department of Medical Mycology, Institute of Dermatology, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing 210042, China; (W.L.); (Q.W.); (X.K.); (G.L.)
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Weida Liu
- Department of Medical Mycology, Institute of Dermatology, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing 210042, China; (W.L.); (Q.W.); (X.K.); (G.L.)
| | - Qiong Wang
- Department of Medical Mycology, Institute of Dermatology, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing 210042, China; (W.L.); (Q.W.); (X.K.); (G.L.)
| | - Xue Kong
- Department of Medical Mycology, Institute of Dermatology, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing 210042, China; (W.L.); (Q.W.); (X.K.); (G.L.)
| | - Guanzhao Liang
- Department of Medical Mycology, Institute of Dermatology, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing 210042, China; (W.L.); (Q.W.); (X.K.); (G.L.)
| | - Santosh Chokkakula
- Department of Microbiology, Chungbuk National University, College of Medicine and Medical Research Institute, Cheongju 28644, Republic of Korea;
| | - Vasundhara Pathakamuri
- Department of Radio-Diagnosis, Sri Venkateshwara Medical College, Tirupathi 517507, India;
| | - Venkatrao Nunna
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA;
| |
Collapse
|
21
|
Onozato R, Miyata J, Asakura T, Namkoong H, Asano K, Hasegawa N, Fukunaga K. Development of allergic bronchopulmonary aspergillosis in a patient with nontuberculous mycobacterial-pulmonary disease successfully treated with dupilumab: A case report and literature review. Respirol Case Rep 2024; 12:e01432. [PMID: 38988827 PMCID: PMC11233258 DOI: 10.1002/rcr2.1432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 06/26/2024] [Indexed: 07/12/2024] Open
Abstract
Pulmonary manifestations in patients with allergic bronchopulmonary aspergillosis (ABPA) and nontuberculous mycobacterial-pulmonary disease (NTM-PD) include bronchiectasis and mucus plugging. A 68-year-old woman, treated with antibiotics and inhaled corticosteroids for NTM-PD and asthma, presented with fever and wheezing. ABPA was diagnosed based on laboratory findings (elevated peripheral blood eosinophil counts and serum total IgE levels and positive Aspergillus-specific IgE and IgG) and imaging observation of a high-attenuation mucus plug. Systemic prednisolone was avoided to prevent NTM-PD progression. Dupilumab, a monoclonal antibody that blocks IL-4/13, was introduced to improve the clinical findings. Herein, we discuss the pathophysiological mechanisms underlying this rare comorbidity.
Collapse
Affiliation(s)
- Ryuta Onozato
- Division of Pulmonary Medicine, Department of Medicine Keio University School of Medicine Tokyo Japan
| | - Jun Miyata
- Division of Pulmonary Medicine, Department of Medicine Keio University School of Medicine Tokyo Japan
| | - Takanori Asakura
- Division of Pulmonary Medicine, Department of Medicine Keio University School of Medicine Tokyo Japan
- Department of Respiratory Medicine Kitasato University Kitasato Institute Hospital Tokyo Japan
- Department of Clinical Medicine (Laboratory of Bioregulatory Medicine) Kitasato University School of Pharmacy Tokyo Japan
| | - Ho Namkoong
- Division of Pulmonary Medicine, Department of Medicine Keio University School of Medicine Tokyo Japan
- Department of Infectious Diseases Keio University School of Medicine Tokyo Japan
| | - Koichiro Asano
- Division of Pulmonary Medicine, Department of Medicine Tokai University School of Medicine Kanagawa Japan
| | - Naoki Hasegawa
- Department of Infectious Diseases Keio University School of Medicine Tokyo Japan
| | - Koichi Fukunaga
- Division of Pulmonary Medicine, Department of Medicine Keio University School of Medicine Tokyo Japan
| |
Collapse
|
22
|
Mac Aogáin M, Dicker AJ, Mertsch P, Chotirmall SH. Infection and the microbiome in bronchiectasis. Eur Respir Rev 2024; 33:240038. [PMID: 38960615 PMCID: PMC11220623 DOI: 10.1183/16000617.0038-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/02/2024] [Indexed: 07/05/2024] Open
Abstract
Bronchiectasis is marked by bronchial dilatation, recurrent infections and significant morbidity, underpinned by a complex interplay between microbial dysbiosis and immune dysregulation. The identification of distinct endophenotypes have refined our understanding of its pathogenesis, including its heterogeneous disease mechanisms that influence treatment and prognosis responses. Next-generation sequencing (NGS) has revolutionised the way we view airway microbiology, allowing insights into the "unculturable". Understanding the bronchiectasis microbiome through targeted amplicon sequencing and/or shotgun metagenomics has provided key information on the interplay of the microbiome and host immunity, a central feature of disease progression. The rapid increase in translational and clinical studies in bronchiectasis now provides scope for the application of precision medicine and a better understanding of the efficacy of interventions aimed at restoring microbial balance and/or modulating immune responses. Holistic integration of these insights is driving an evolving paradigm shift in our understanding of bronchiectasis, which includes the critical role of the microbiome and its unique interplay with clinical, inflammatory, immunological and metabolic factors. Here, we review the current state of infection and the microbiome in bronchiectasis and provide views on the future directions in this field.
Collapse
Affiliation(s)
- Micheál Mac Aogáin
- Biochemical Genetics Laboratory, Department of Biochemistry, St. James's Hospital, Dublin, Ireland
- Clinical Biochemistry Unit, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Alison J Dicker
- Respiratory Research Group, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Pontus Mertsch
- Department of Medicine V, LMU University Hospital, LMU Munich, Comprehensive Pneumology Center (CPC), Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Sanjay H Chotirmall
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Respiratory and Critical Care Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| |
Collapse
|
23
|
Martins Oliveira-Brito PK, de Campos GY, Guimarães JG, Machado MP, Serafim LC, Lazo Chica JE, Roque-Barreira MC, da Silva TA. Adjuvant ArtinM favored the host immunity against Cryptococcus gattii infection in C57BL/6 mice. Immunotherapy 2024; 16:733-748. [PMID: 38940276 PMCID: PMC11421300 DOI: 10.1080/1750743x.2024.2360384] [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: 01/15/2024] [Accepted: 05/22/2024] [Indexed: 06/29/2024] Open
Abstract
Aim: Cryptococcus gattii causes a severe fungal infection with high mortality rate among immunosuppressed and immunocompetent individuals. Due to limitation of current antifungal treatment, new immunotherapeutic approaches are explored.Methods: This study investigated an immunization strategy utilizing heat-inactivated C. gattii with ArtinM as an adjuvant. C57BL/6 mice were intranasally immunized with heat-killed C. gattii and ArtinM was administrated either before immunization or along with HK-C. gattii. Mice were infected with C. gattii and the efficacy of the immunization protocol was evaluated.Results: Mice that received ArtinM exhibited increased levels of IL-10 and relative expression of IL-23 in the lungs, reduced fungal burden and preserved tissue integrity post-infection.Conclusion: Adjuvant ArtinM improved immunization against C. gattii infection in C57BL/6 mice.
Collapse
Affiliation(s)
- Patrícia Kellen Martins Oliveira-Brito
- Department of Cell & Molecular Biology & Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Gabriela Yamazaki de Campos
- Department of Cell & Molecular Biology & Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Júlia Garcia Guimarães
- Department of Cell & Molecular Biology & Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Michele Procópio Machado
- Department of Cell & Molecular Biology & Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Letícia Costa Serafim
- Microbiology Postgraduate Program of the Microbiology Department of the Biomedical Sciences Institute (ICB) of University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Javier Emílio Lazo Chica
- Institute of Natural & Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, Brazil
| | - Maria Cristina Roque-Barreira
- Department of Cell & Molecular Biology & Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Thiago Aparecido da Silva
- Department of Cell & Molecular Biology & Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Clinical Hematology Lab, Department of Clinical Analysis, School of Pharmaceutical Sciences in Araraquara (FCFAR), Sao Paulo State University (UNESP), Araraquara, São Paulo, Brazil
- National Institute of Science & Technology in Human Pathogenic Fungi, School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirão Preto, São Paulo,Brazil
| |
Collapse
|
24
|
Neoh CF, Chen SCA, Lanternier F, Tio SY, Halliday CL, Kidd SE, Kong DCM, Meyer W, Hoenigl M, Slavin MA. Scedosporiosis and lomentosporiosis: modern perspectives on these difficult-to-treat rare mold infections. Clin Microbiol Rev 2024; 37:e0000423. [PMID: 38551323 PMCID: PMC11237582 DOI: 10.1128/cmr.00004-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024] Open
Abstract
SUMMARYAlthough Scedosporium species and Lomentospora prolificans are uncommon causes of invasive fungal diseases (IFDs), these infections are associated with high mortality and are costly to treat with a limited armamentarium of antifungal drugs. In light of recent advances, including in the area of new antifungals, the present review provides a timely and updated overview of these IFDs, with a focus on the taxonomy, clinical epidemiology, pathogenesis and host immune response, disease manifestations, diagnosis, antifungal susceptibility, and treatment. An expansion of hosts at risk for these difficult-to-treat infections has emerged over the last two decades given the increased use of, and broader population treated with, immunomodulatory and targeted molecular agents as well as wider adoption of antifungal prophylaxis. Clinical presentations differ not only between genera but also across the different Scedosporium species. L. prolificans is intrinsically resistant to most currently available antifungal agents, and the prognosis of immunocompromised patients with lomentosporiosis is poor. Development of, and improved access to, diagnostic modalities for early detection of these rare mold infections is paramount for timely targeted antifungal therapy and surgery if indicated. New antifungal agents (e.g., olorofim, fosmanogepix) with novel mechanisms of action and less cross-resistance to existing classes, availability of formulations for oral administration, and fewer drug-drug interactions are now in late-stage clinical trials, and soon, could extend options to treat scedosporiosis/lomentosporiosis. Much work remains to increase our understanding of these infections, especially in the pediatric setting. Knowledge gaps for future research are highlighted in the review.
Collapse
Affiliation(s)
- Chin Fen Neoh
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Sharon C-A Chen
- Centre for Infectious Diseases and Microbiology Laboratory Services, New South Wales Health Pathology, Westmead Hospital, Sydney, Australia
- The University of Sydney, Sydney, Australia
- Department of Infectious Diseases, Westmead Hospital, Sydney, Australia
| | - Fanny Lanternier
- Service de Maladies Infectieuses et Tropicales, Hôpital universitaire Necker-Enfants malades, Paris, France
- National Reference Center for Invasive Mycoses and Antifungals, Translational Mycology research group, Mycology Department, Institut Pasteur, Université Paris Cité, Paris, France
| | - Shio Yen Tio
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Catriona L Halliday
- Centre for Infectious Diseases and Microbiology Laboratory Services, New South Wales Health Pathology, Westmead Hospital, Sydney, Australia
| | - Sarah E Kidd
- National Mycology Reference Centre, SA Pathology, Adelaide, Australia
- School of Biological Sciences, Faculty of Sciences, University of Adelaide, Adelaide, Australia
| | - David C M Kong
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Australia
- The National Centre for Antimicrobial Stewardship, The Peter Doherty Institute for Infections and Immunity, Melbourne, Australia
- Centre for Medicine Use and Safety, Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Melbourne, Australia
- School of Medicine, Deakin University, Waurn Ponds, Geelong, Australia
| | - Wieland Meyer
- The University of Sydney, Sydney, Australia
- Westerdijk Fungal Biodiversity Institute, Utrecht, the Netherlands
| | - Martin Hoenigl
- Division of Infectious Diseases, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Translational Medical Mycology Research Group, ECMM Excellence Center for Clinical Mycology, Medical University of Graz, Graz, Austria
| | - Monica A Slavin
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
25
|
Shin SH, Ye MK, Lee DW, Choi MH. Impact of Zinc Oxide on the Development of Aspergillus-Induced Maxillary Sinusitis Rabbit Model. Curr Issues Mol Biol 2024; 46:5712-5723. [PMID: 38921013 PMCID: PMC11202535 DOI: 10.3390/cimb46060342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 05/29/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024] Open
Abstract
Aspergillus fumigatus is commonly found in the airway and is associated with airway inflammatory diseases. Zinc oxide (ZO) is known to be an essential microelement that facilitates fungal survival, growth, and proliferation. This study aimed to investigate the impact of ZO on A. fumigatus-induced fungal sinusitis in rabbits. Twenty-eight New Zealand white rabbits were divided into four groups for this study. Group 1 (6 sides) was treated with intramaxillary phosphate buffer saline (PBS) served as the negative control, Group 2 (6 sides) received intramaxillary PBS and ZO, Group 3 (8 sides) was treated with intramaxillary A. fumigatus alone, and Group 4 (8 sides) treated with intramaxillary A. fumigatus with ZO. After 4 and 12 weeks, sinus mucosal cytokine and transcription factor expressions were determined. A histological analysis was performed to determine inflammatory cell infiltration, number of secretory cells, and mucosal thickness. Fungal biofilm formation was determined using confocal laser microscopy. The intramaxillary instillation of A. fumigatus conidia led to an increase in protein and mRNA expression of interleukin (IL)-1β and IL-8 in the maxillary sinus mucosa. They were associated with mitogen-activated protein kinase and activator protein-1. Furthermore, intramaxillary instillation of fungal conidia resulted in significant enhancement of inflammatory cell infiltration, epithelial thickening, and fungal biofilm formation. However, intramaxillary ZO did not have a significant impact on A. fumigatus-induced cytokine protein and mRNA expression, and inflammatory cell infiltration and epithelial thickness in sinonasal mucosa. While intramaxillary instillation of A. fumigatus increased mucosal inflammation, cytokine production, and biofilm formation, the intramaxillary application of ZO did not have a significant influence on inflammation in the maxillary sinus mucosa.
Collapse
Affiliation(s)
- Seung-Heon Shin
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea; (M.-K.Y.); (D.-W.L.); (M.-H.C.)
| | | | | | | |
Collapse
|
26
|
Lyu S, Zhang T, Peng P, Cao D, Ma L, Yu Y, Dong Y, Qi X, Wei C. Involvement of cGAS/STING Signaling in the Pathogenesis of Candida albicans Keratitis: Insights From Genetic and Pharmacological Approaches. Invest Ophthalmol Vis Sci 2024; 65:13. [PMID: 38848078 PMCID: PMC11166223 DOI: 10.1167/iovs.65.6.13] [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: 12/27/2023] [Accepted: 05/14/2024] [Indexed: 06/13/2024] Open
Abstract
Purpose Fungal keratitis (FK) is an invasive corneal infection associated with significant risk to vision. Although the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) signaling pathway has been recognized for its role in defending against viral infections, its involvement in FK still remains largely unclear. This study sought to elucidate the contribution of the cGAS/STING signaling pathway to the pathogenesis of FK. Methods The expression of cGAS/STING signaling components was assessed in a murine model of Candida albicans keratitis through RNA sequencing, western blot analysis, immunofluorescence staining, and real-time PCR. Both genetic (utilizing Sting1gt/gt mice) and pharmacological (using C176) interventions were employed to inhibit STING activity, allowing for the evaluation of resultant pathogenic alterations in FK using slit-lamp examination, clinical scoring, hematoxylin and eosin (H&E) staining, fungal culture, and RNA sequencing. Subconjunctival administration of the NOD-like receptor protein 3 (NLRP3) inflammasome inhibitor MCC950 was performed to evaluate FK manifestations following STING activity blockade. Furthermore, the impact of the STING agonist diABZI on FK progression was investigated. Results Compared to uninfected corneas, those infected with C. albicans exhibited increased expression of cGAS/STING signaling components, as well as its elevated activity. Inhibiting cGAS/STING signaling exacerbated the advancement of FK, as evidenced by elevated clinical scores, augmented fungal load, and heightened inflammatory response, including NLRP3 inflammasome activation and pyroptosis. Pharmacological inhibition of the NLRP3 inflammasome effectively mitigated the exacerbated FK by suppressing STING activity. Conversely, pre-activation of STING exacerbated FK progression compared to the PBS control, characterized by increased fungal burden and reinforced inflammatory infiltration. Conclusions This study demonstrates the essential role of the cGAS/STING signaling pathway in FK pathogenesis and highlights the necessity of its proper activation for the host against FK.
Collapse
Affiliation(s)
- Shanmei Lyu
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Ting Zhang
- Eye Hospital of Shandong First Medical University, Eye Institute of Shandong First Medical University, Jinan, Shandong, China
| | - Peng Peng
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Dingwen Cao
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Li Ma
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Yang Yu
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Yanling Dong
- Qingdao Eye Hospital of Shandong First Medical University, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Xiaolin Qi
- Eye Hospital of Shandong First Medical University, Eye Institute of Shandong First Medical University, Jinan, Shandong, China
- School of Ophthalmology, Shandong First Medical University, Jinan, Shandong, China
| | - Chao Wei
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| |
Collapse
|
27
|
Zhang J, Liu Y, Zhi X, Xu L, Tao J, Cui D, Liu TF. Tryptophan catabolism via the kynurenine pathway regulates infection and inflammation: from mechanisms to biomarkers and therapies. Inflamm Res 2024; 73:979-996. [PMID: 38592457 DOI: 10.1007/s00011-024-01878-5] [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: 12/06/2023] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND L-Tryptophan (L-Trp), an essential amino acid, is the only amino acid whose level is regulated specifically by immune signals. Most proportions of Trp are catabolized via the kynurenine (Kyn) pathway (KP) which has evolved to align the food availability and environmental stimulation with the host pathophysiology and behavior. Especially, the KP plays an indispensable role in balancing the immune activation and tolerance in response to pathogens. SCOPE OF REVIEW In this review, we elucidate the underlying immunological regulatory network of Trp and its KP-dependent catabolites in the pathophysiological conditions by participating in multiple signaling pathways. Furthermore, the KP-based regulatory roles, biomarkers, and therapeutic strategies in pathologically immune disorders are summarized covering from acute to chronic infection and inflammation. MAJOR CONCLUSIONS The immunosuppressive effects dominate the functions of KP induced-Trp depletion and KP-produced metabolites during infection and inflammation. However, the extending minor branches from the KP are not confined to the immune tolerance, instead they go forward to various functions according to the specific condition. Nevertheless, persistent efforts should be made before the clinical use of KP-based strategies to monitor and cure infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Jingpu Zhang
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Highway, Shanghai, 201508, People's Republic of China.
| | - Yanlei Liu
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Xiao Zhi
- Shanghai Institute of Virology, Shanghai Jiao Tong University School of Medicine, 227 South Chongqing Road, Shanghai, 200025, People's Republic of China
| | - Li Xu
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Highway, Shanghai, 201508, People's Republic of China
| | - Jie Tao
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Highway, Shanghai, 201508, People's Republic of China
| | - Daxiang Cui
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Tie Fu Liu
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Highway, Shanghai, 201508, People's Republic of China.
| |
Collapse
|
28
|
Wrońska AK, Kaczmarek A, Sobich J, Boguś MI. The effect of infection with the entomopathogenic fungus Conidiobolus coronatus (Entomopthorales) on eighteen cytokine-like proteins in Galleria mellonella (Lepidoptera) larvae. Front Immunol 2024; 15:1385863. [PMID: 38774871 PMCID: PMC11106378 DOI: 10.3389/fimmu.2024.1385863] [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: 02/13/2024] [Accepted: 04/16/2024] [Indexed: 05/24/2024] Open
Abstract
Background In response to the replace mammal research models with insects in preliminary immunological studies, interest has grown in invertebrate defense systems. The immunological response is regulated by cytokines; however, while their role in mammals is well understood, little is known of their function in insects. A suitable target for studies into insect immunology is Galleria mellonella (Lepidoptera), the wax moth: a common host for human fungal and bacterial pathogens. G. mellonella is also a perfect subject for studies into the presence of cytokine-like proteins. Specific objectives The main goal of present research was detection in insect immunocompetent cells the 18 mammalian cytokines (IL-1α, IL-1β, IL-2, IL-3, IL-6, IL-7, IL-8, IL-12, IL-13, IL-15, IL-17, IL-19, IFN-γ, TNF-α, TNF-β, GM-CSF, M-CSF, G-CSF), which play important role in immunological response and indication how their level change after fungal infection. Methodology The changes of cytokine-like proteins level were detected in hemocytes taken from G. mellonella larvae infected with entomopathogenic fungus, C. coronatus. The presence of cytokine-proteins was confirmed with using fluorescence microscopy (in cultured hemocytes) and flow cytometry (in freshly collected hemolymph). The ELISA test was used to detect changes in concentration of examined cytokine-like proteins. Results Our findings indicated the presence of eighteen cytokine-like molecules in G. mellonella hemocytes during infection with C. coronatus. The hemocytes taken from infected larvae demonstrated higher fluorescence intensity for six cytokine-like proteins (GM-CSF, M-CSF, IL-3, IL-15, IL-1β and IL-19) compared to untreated controls. ELISA test indicated significantly higher IL-3 and IL-15. M-CSF, IL-1α and IL-19 concentration in the hemolymph after fungal infection, and significantly lower TNF-β and G-CSF. Conclusions Our findings confirm that the selected cytokine-like molecules are present in insect hemocytes and that their concentrations change after fungal infection, which might suggest that they play a role in the anti-fungal immunological response.
Collapse
Affiliation(s)
| | - Agata Kaczmarek
- Museum and Institute of Zoology, Polish Academy of Science, Warsaw, Poland
| | - Justyna Sobich
- Museum and Institute of Zoology, Polish Academy of Science, Warsaw, Poland
- Dioscuri Centre for RNA-Protein Interactions in Human Health and Disease, International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | | |
Collapse
|
29
|
Nenciarini S, Renzi S, di Paola M, Meriggi N, Cavalieri D. The yeast-human coevolution: Fungal transition from passengers, colonizers, and invaders. WIREs Mech Dis 2024; 16:e1639. [PMID: 38146626 DOI: 10.1002/wsbm.1639] [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: 05/19/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/27/2023]
Abstract
Fungi are the cause of more than a billion infections in humans every year, although their interactions with the host are still neglected compared to bacteria. Major systemic fungal infections are very unusual in the healthy population, due to the long history of coevolution with the human host. Humans are routinely exposed to environmental fungi and can host a commensal mycobiota, which is increasingly considered as a key player in health and disease. Here, we review the current knowledge on host-fungi coevolution and the factors that regulate their interaction. On one hand, fungi have learned to survive and inhabit the host organisms as a natural ecosystem, on the other hand, the host immune system finely tunes the response toward fungi. In turn, recognition of fungi as commensals or pathogens regulates the host immune balance in health and disease. In the human gut ecosystem, yeasts provide a fingerprint of the transient microbiota. Their status as passengers or colonizers is related to the integrity of the gut barrier and the risk of multiple disorders. Thus, the study of this less known component of the microbiota could unravel the rules of the transition from passengers to colonizers and invaders, as well as their dependence on the innate component of the host's immune response. This article is categorized under: Infectious Diseases > Environmental Factors Immune System Diseases > Environmental Factors Infectious Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
| | - Sonia Renzi
- Department of Biology, University of Florence, Florence, Italy
| | - Monica di Paola
- Department of Biology, University of Florence, Florence, Italy
| | - Niccolò Meriggi
- Department of Biology, University of Florence, Florence, Italy
| | | |
Collapse
|
30
|
Kudari SM, Rani S, Mendiratta V, Sonker S, Rawat D, Xess I, Singh S. Medicopsis romeroi: A causative agent of subcutaneous phaeohyphomycosis in a diabetic patient. Indian J Dermatol Venereol Leprol 2024; 0:1-2. [PMID: 38841965 DOI: 10.25259/ijdvl_675_2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 12/12/2023] [Indexed: 06/07/2024]
Affiliation(s)
- Shankar Mohan Kudari
- Department of Dermatology, Lady Hardinge Medical College & Smt S. K. Hospital, New Delhi, India
| | - Seema Rani
- Department of Dermatology, Lady Hardinge Medical College & Smt S. K. Hospital, New Delhi, India
| | - Vibhu Mendiratta
- Department of Dermatology, Lady Hardinge Medical College & Smt S. K. Hospital, New Delhi, India
| | - Sneha Sonker
- Department of Microbiology, Lady Hardinge Medical College & Smt S. K. Hospital, New Delhi, India
| | - Deepti Rawat
- Department of Microbiology, Lady Hardinge Medical College & Smt S. K. Hospital, New Delhi, India
| | | | - Smita Singh
- Department of Pathology, Lady Hardinge Medical College & Smt S.K. Hospital, Delhi, India
| |
Collapse
|
31
|
Arunan B, Talukdar D, Swain S, Varadarajan A, Sarda R, Singh G, Nischal N, Soneja M, Bakshi S, Jana P, Tanwar S, Sikka K, Verma H, Subramanian A, Xess I, Wig N, Das B, Ray A. Metagenomic insights into fungal community composition of the nasopharyngeal region of COVID-19 associated mucormycosis patients from India. J Med Virol 2024; 96:e29601. [PMID: 38597375 DOI: 10.1002/jmv.29601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/22/2024] [Accepted: 04/01/2024] [Indexed: 04/11/2024]
Abstract
Coronavirus disease 2019 (COVID-19) associated mucormycosis (CAM) was reported predominantly from India during the second wave of COVID-19 and has a high mortality rate. The present study aims to understand the fungal community composition of the nasopharyngeal region of CAM-infected individuals and compare it with severe COVID-19 patients and healthy controls. The fungal community composition was decoded by analyzing the sequence homology of the internal transcribed spacer-2-(ITS-2) region of metagenomic DNA extracted from the upper respiratory samples. The alpha-diversity indices were found to be significantly altered in CAM patients (p < 0.05). Interestingly, a higher abundance of Candida africana, Candida haemuloni, Starmerella floris, and Starmerella lactiscondensi was observed exclusively in CAM patients. The interindividual changes in mycobiome composition were well supported by beta-diversity analysis (p < 0.05). The current study provides insights into the dysbiosis of the nasal mycobiome during CAM infection. In conclusion, our study shows that severe COVID-19 and CAM are associated with alteration in mycobiome as compared to healthy controls. However, the sequential alteration in the fungal flora which ultimately leads to the development of CAM needs to be addressed by future studies.
Collapse
Affiliation(s)
| | - Daizee Talukdar
- Functional Genomics Laboratory, BRIC-THSTI, Faridabad, Haryana, India
| | - Satish Swain
- Department of Medicine, AIIMS, New Delhi, Delhi, India
| | | | - Radhika Sarda
- Department of Medicine, AIIMS, New Delhi, Delhi, India
| | | | | | - Manish Soneja
- Department of Medicine, AIIMS, New Delhi, Delhi, India
| | - Susmita Bakshi
- Functional Genomics Laboratory, BRIC-THSTI, Faridabad, Haryana, India
| | - Pradipta Jana
- Functional Genomics Laboratory, BRIC-THSTI, Faridabad, Haryana, India
| | - Subhash Tanwar
- Functional Genomics Laboratory, BRIC-THSTI, Faridabad, Haryana, India
| | - Kapil Sikka
- Department of Otorhinolaryngology, AIIMS, New Delhi, Delhi, India
| | - Hitesh Verma
- Department of Otorhinolaryngology, AIIMS, New Delhi, Delhi, India
| | | | | | - Naveet Wig
- Department of Medicine, AIIMS, New Delhi, Delhi, India
| | - Bhabatosh Das
- Functional Genomics Laboratory, BRIC-THSTI, Faridabad, Haryana, India
| | - Animesh Ray
- Department of Medicine, AIIMS, New Delhi, Delhi, India
| |
Collapse
|
32
|
Kumar V, Huang J, Dong Y, Hao GF. Targeting Fks1 proteins for novel antifungal drug discovery. Trends Pharmacol Sci 2024; 45:366-384. [PMID: 38493014 DOI: 10.1016/j.tips.2024.02.007] [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: 01/09/2024] [Revised: 01/26/2024] [Accepted: 02/26/2024] [Indexed: 03/18/2024]
Abstract
Fungal infections are a major threat to human health. The limited availability of antifungal drugs, the emergence of drug resistance, and a growing susceptible population highlight the critical need for novel antifungal agents. The enzymes involved in fungal cell wall synthesis offer potential targets for antifungal drug development. Recent studies have enhanced our focus on the enzyme Fks1, which synthesizes β-1,3-glucan, a critical component of the cell wall. These studies provide a deeper understanding of Fks1's function in cell wall biosynthesis, pathogenicity, structural biology, evolutionary conservation across fungi, and interaction with current antifungal drugs. Here, we discuss the role of Fks1 in the survival and adaptation of fungi, guided by insights from evolutionary and structural analyses. Furthermore, we delve into the dynamics of Fks1 modulation with novel antifungal strategies and assess its potential as an antifungal drug target.
Collapse
Affiliation(s)
- Vinit Kumar
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University, Guiyang 550025, PR China; BMLT, Markham College of Commerce, Vinoba Bhave University, Hazaribagh, Jharkhand 825301, India
| | - Juan Huang
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, PR China
| | - Yawen Dong
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, PR China.
| | - Ge-Fei Hao
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University, Guiyang 550025, PR China; National Key Laboratory of Green Pesticide, Central China Normal University, Wuhan 430079, PR China.
| |
Collapse
|
33
|
Chiu CY, John TM, Matsuo T, Wurster S, Hicklen RS, Khattak RR, Ariza-Heredia EJ, Bose P, Kontoyiannis DP. Disseminated Histoplasmosis in a Patient with Myelofibrosis on Ruxolitinib: A Case Report and Review of the Literature on Ruxolitinib-Associated Invasive Fungal Infections. J Fungi (Basel) 2024; 10:264. [PMID: 38667935 PMCID: PMC11051496 DOI: 10.3390/jof10040264] [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: 02/24/2024] [Revised: 03/24/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
Ruxolitinib, a selective inhibitor of Janus kinases, is a standard treatment for intermediate/high-risk myelofibrosis (MF) but is associated with a predisposition to opportunistic infections, especially herpes zoster. However, the incidence and characteristics of invasive fungal infections (IFIs) in these patients remain uncertain. In this report, we present the case of a 59-year-old woman with MF who developed disseminated histoplasmosis after seven months of ruxolitinib use. The patient clinically improved after ten weeks of combined amphotericin B and azole therapy, and ruxolitinib was discontinued. Later, the patient received fedratinib, a relatively JAK2-selective inhibitor, without relapse of histoplasmosis. We also reviewed the literature on published cases of proven IFIs in patients with MF who received ruxolitinib. Including ours, we identified 28 such cases, most commonly due to Cryptococcus species (46%). IFIs were most commonly disseminated (39%), followed by localized lung (21%) infections. Although uncommon, a high index of suspicion for opportunistic IFIs is needed in patients receiving JAK inhibitors. Furthermore, the paucity of data regarding the optimal management of IFIs in patients treated with JAK inhibitors underscore the need for well-designed studies to evaluate the epidemiology, pathobiology, early diagnosis, and multimodal therapy of IFIs in patients with hematological malignancies receiving targeted therapies.
Collapse
Affiliation(s)
- Chia-Yu Chiu
- Department of Infectious Diseases, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.-Y.C.); (T.M.J.); (T.M.); (S.W.); (R.R.K.); (E.J.A.-H.)
| | - Teny M. John
- Department of Infectious Diseases, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.-Y.C.); (T.M.J.); (T.M.); (S.W.); (R.R.K.); (E.J.A.-H.)
| | - Takahiro Matsuo
- Department of Infectious Diseases, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.-Y.C.); (T.M.J.); (T.M.); (S.W.); (R.R.K.); (E.J.A.-H.)
| | - Sebastian Wurster
- Department of Infectious Diseases, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.-Y.C.); (T.M.J.); (T.M.); (S.W.); (R.R.K.); (E.J.A.-H.)
| | - Rachel S. Hicklen
- Research Medical Library, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Raihaan Riaz Khattak
- Department of Infectious Diseases, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.-Y.C.); (T.M.J.); (T.M.); (S.W.); (R.R.K.); (E.J.A.-H.)
| | - Ella J. Ariza-Heredia
- Department of Infectious Diseases, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.-Y.C.); (T.M.J.); (T.M.); (S.W.); (R.R.K.); (E.J.A.-H.)
| | - Prithviraj Bose
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Dimitrios P. Kontoyiannis
- Department of Infectious Diseases, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.-Y.C.); (T.M.J.); (T.M.); (S.W.); (R.R.K.); (E.J.A.-H.)
| |
Collapse
|
34
|
Rinker DC, Sauters TJC, Steffen K, Gumilang A, Raja HA, Rangel-Grimaldo M, Pinzan CF, de Castro PA, dos Reis TF, Delbaje E, Houbraken J, Goldman GH, Oberlies NH, Rokas A. Strain heterogeneity in a non-pathogenic fungus highlights factors contributing to virulence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.583994. [PMID: 38496489 PMCID: PMC10942418 DOI: 10.1101/2024.03.08.583994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Fungal pathogens exhibit extensive strain heterogeneity, including variation in virulence. Whether closely related non-pathogenic species also exhibit strain heterogeneity remains unknown. Here, we comprehensively characterized the pathogenic potentials (i.e., the ability to cause morbidity and mortality) of 16 diverse strains of Aspergillus fischeri, a non-pathogenic close relative of the major pathogen Aspergillus fumigatus. In vitro immune response assays and in vivo virulence assays using a mouse model of pulmonary aspergillosis showed that A. fischeri strains varied widely in their pathogenic potential. Furthermore, pangenome analyses suggest that A. fischeri genomic and phenotypic diversity is even greater. Genomic, transcriptomic, and metabolomic profiling identified several pathways and secondary metabolites associated with variation in virulence. Notably, strain virulence was associated with the simultaneous presence of the secondary metabolites hexadehydroastechrome and gliotoxin. We submit that examining the pathogenic potentials of non-pathogenic close relatives is key for understanding the origins of fungal pathogenicity.
Collapse
Affiliation(s)
- David C. Rinker
- Department of Biological Sciences and Evolutionary Studies Initiative, Vanderbilt University, Nashville, Tennessee, USA
| | - Thomas J. C. Sauters
- Department of Biological Sciences and Evolutionary Studies Initiative, Vanderbilt University, Nashville, Tennessee, USA
| | - Karin Steffen
- Department of Biological Sciences and Evolutionary Studies Initiative, Vanderbilt University, Nashville, Tennessee, USA
| | - Adiyantara Gumilang
- Department of Biological Sciences and Evolutionary Studies Initiative, Vanderbilt University, Nashville, Tennessee, USA
| | - Huzefa A. Raja
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina, USA
| | - Manuel Rangel-Grimaldo
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina, USA
| | - Camila Figueiredo Pinzan
- Faculdade de Ciencias Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Patrícia Alves de Castro
- Faculdade de Ciencias Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Thaila Fernanda dos Reis
- Faculdade de Ciencias Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Endrews Delbaje
- Faculdade de Ciencias Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Jos Houbraken
- Food and Indoor Mycology, Westerdijk Fungal Biodiversity Institute, Utrecht, The Netherlands
| | - Gustavo H. Goldman
- Faculdade de Ciencias Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Nicholas H. Oberlies
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina, USA
| | - Antonis Rokas
- Department of Biological Sciences and Evolutionary Studies Initiative, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
35
|
Hansakon A, Angkasekwinai P. Murine Models of Cryptococcus Infection. Curr Protoc 2024; 4:e1001. [PMID: 38456766 DOI: 10.1002/cpz1.1001] [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: 03/09/2024]
Abstract
Cryptococcus is recognized as one of the emerging fungal pathogens that have major impact on diverse populations worldwide. Because of the high mortality rate and limited antifungal therapy options, there is an urgent need to understand the impact of dynamic processes between fungal pathogens and hosts that influence cryptococcal pathogenesis and disease outcomes. With known common limitations in human studies, experimental murine cryptococcosis models that can recapitulate human disease provide a valuable tool for studying fungal virulence and the host interaction, leading to development of better treatment strategies. Infection with Cryptococcus in mice via intranasal inhalation is mostly used because it is noninvasive and considered to be the most common mode of infection, strongly correlating with cryptococcal disease in humans. The protocols described in this article provide the procedures of establishing a murine model of Cryptococcus infection by intranasal inhalation and assessing the host immune response and disease progression during Cryptococcus infection. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Murine model of pulmonary cryptococcal infection via intranasal inhalation Basic Protocol 2: Assessment of the pulmonary immune response during Cryptococcus infection Support Protocol: Evaluation of pulmonary gene expression by real-time PCR Basic Protocol 3: Enumeration of survival rate and organ fungal burden.
Collapse
Affiliation(s)
- Adithap Hansakon
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Pornpimon Angkasekwinai
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, Thailand
| |
Collapse
|
36
|
Rodriguez KA, Gurung M, Talatala R, Rearick JR, Ruebel ML, Stephens KE, Yeruva L. The Role of Early Life Gut Mycobiome on Child Health. Adv Nutr 2024; 15:100185. [PMID: 38311313 PMCID: PMC10907404 DOI: 10.1016/j.advnut.2024.100185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/10/2024] Open
Abstract
The human gut microbiota is composed of bacteria (microbiota or microbiome), fungi (mycobiome), viruses, and archaea, but most of the research is primarily focused on the bacterial component of this ecosystem. Besides bacteria, fungi have been shown to play a role in host health and physiologic functions. However, studies on mycobiota composition during infancy, the factors that might shape infant gut mycobiota, and implications to child health and development are limited. In this review, we discuss the factors likely shaping gut mycobiota, interkingdom interactions, and associations with child health outcomes and highlight the gaps in our current knowledge of this ecosystem.
Collapse
Affiliation(s)
- Kayleigh Amber Rodriguez
- Arkansas Children's Research Institute, Little Rock, AR, United States; University of Arkansas for Medical Sciences, Department of Pediatrics, Division of Infectious Diseases, Little Rock, AR, United States
| | - Manoj Gurung
- Microbiome and Metabolism Research Unit, United States Department of Agriculture, Agriculture Research Service, Little Rock, AR, United States; Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - Rachelanne Talatala
- Microbiome and Metabolism Research Unit, United States Department of Agriculture, Agriculture Research Service, Little Rock, AR, United States
| | - Jolene R Rearick
- Microbiome and Metabolism Research Unit, United States Department of Agriculture, Agriculture Research Service, Little Rock, AR, United States; Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - Meghan L Ruebel
- Microbiome and Metabolism Research Unit, United States Department of Agriculture, Agriculture Research Service, Little Rock, AR, United States; Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - Kimberly E Stephens
- Arkansas Children's Research Institute, Little Rock, AR, United States; University of Arkansas for Medical Sciences, Department of Pediatrics, Division of Infectious Diseases, Little Rock, AR, United States.
| | - Laxmi Yeruva
- Microbiome and Metabolism Research Unit, United States Department of Agriculture, Agriculture Research Service, Little Rock, AR, United States; Arkansas Children's Nutrition Center, Little Rock, AR, United States.
| |
Collapse
|
37
|
Zhang Y, Zhao M, He J, Chen L, Wang W. In vitro and in vivo immunomodulatory activity of acetylated polysaccharides from Cyclocarya paliurus leaves. Int J Biol Macromol 2024; 259:129174. [PMID: 38181912 DOI: 10.1016/j.ijbiomac.2023.129174] [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: 04/24/2023] [Revised: 12/04/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024]
Abstract
In this study, we aimed to investigate the immunomodulatory effects of polysaccharides from Cyclocarya paliurus leaves after acetylation modification (Ac-CPP0.1) on dendritic cells (DCs) and immunosuppressed mice. In vitro, Ac-CPP0.1 promoted phenotypic and functional maturation of DCs. Specifically, it increased the expression of costimulatory molecules (CD80, CD86, and MHC II) and the secretion of cytokines (TNF-α, IL-6, IL-1β, IL-10, IL-12p70) of DCs. In vivo, Ac-CPP0.1 significantly improved immunosuppression of mice, which was manifested by increased body weight and immune organ index, up-regulated cytokines (IL-4, IL-17, TGF-β3, and TNF-α), and restored short-chain fatty acid (SCFAs) levels of intestinal. The immunoactivation of Ac-CPP0.1 in DCs and in mice is linked to the activation of the TLR4/NF-κB signaling pathway. Furthermore, Ac-CPP0.1 reversed intestinal flora imbalance caused by cyclophosphamide. At the species level, Ac-CPP0.1 increased the abundance of unclassified_Muribaculaceae, unclassified_Desulfovibrio, Bacteroides_acidifaciens and Faecalibaculum_rodentium, decreased the level of Lactobacillus_johnsonii, unclassified_g_Staphylococcus and Staphylococcus_nepalensis. In summary, Ac-CPP0.1 has considerable immunomodulatory potential, which is beneficial to the future utilization and development of Cyclocarya paliurus.
Collapse
Affiliation(s)
- Yang Zhang
- Jiangxi Province Key Laboratory of Animal Nutrition, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| | - Meng Zhao
- Key Lab for Agro-product Processing and Quality Control of Nanchang City, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Jing He
- Key Lab for Agro-product Processing and Quality Control of Nanchang City, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Lingli Chen
- Key Lab for Agro-product Processing and Quality Control of Nanchang City, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Wenjun Wang
- Key Lab for Agro-product Processing and Quality Control of Nanchang City, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China.
| |
Collapse
|
38
|
Schwarz C, Eschenhagen PN, Mainz JG, Schmidergall T, Schuette H, Romanowska E. Pulmonary Aspergillosis in People with Cystic Fibrosis. Semin Respir Crit Care Med 2024; 45:128-140. [PMID: 38286138 DOI: 10.1055/s-0043-1777267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2024]
Abstract
In the last decade, fungal respiratory diseases have been increasingly investigated for their impact on the clinical course of people with cystic fibrosis (CF), with a particular focus on infections caused by Aspergillus spp. The most common organisms from this genus detected from respiratory cultures are Aspergillus fumigatus and Aspergillus terreus, followed by Aspergillus flavus, Aspergillus niger, and Aspergillus nidulans. These species have been identified to be both chronic colonizers and sources of active infection and may negatively impact lung function in people with CF. This review article discusses definitions of aspergillosis, challenges in clinical practice, and current literature available for laboratory findings, clinical diagnosis, and treatment options for pulmonary diseases caused by Aspergillus spp. in people with CF.
Collapse
Affiliation(s)
- C Schwarz
- HMU-Health and Medical University, Potsdam, Germany
- Division Cystic Fibrosis, Clinic Westbrandenburg, Potsdam, Germany
| | - P N Eschenhagen
- HMU-Health and Medical University, Potsdam, Germany
- Division Cystic Fibrosis, Clinic Westbrandenburg, Potsdam, Germany
| | - J G Mainz
- Department of Paediatric Pneumology, Allergology, Cystic Fibrosis Center, Klinikum Westbrandenburg, Brandenburg a. d. Havel, Germany
- University Hospital of the Brandenburg Medical School, Brandenburg a. d. Havel, Germany
| | - T Schmidergall
- HMU-Health and Medical University, Potsdam, Germany
- Division Cystic Fibrosis, Clinic Westbrandenburg, Potsdam, Germany
| | - H Schuette
- Pneumology and Respiratory Medicine, Ernst von Bergmann Klinikum, Potsdam, Germany
| | - E Romanowska
- HMU-Health and Medical University, Potsdam, Germany
- Division Cystic Fibrosis, Clinic Westbrandenburg, Potsdam, Germany
| |
Collapse
|
39
|
Janssens I, Lambrecht BN, Van Braeckel E. Aspergillus and the Lung. Semin Respir Crit Care Med 2024; 45:3-20. [PMID: 38286136 PMCID: PMC10857890 DOI: 10.1055/s-0043-1777259] [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: 01/31/2024]
Abstract
The filamentous fungus Aspergillus causes a wide spectrum of diseases in the human lung, with Aspergillus fumigatus being the most pathogenic and allergenic subspecies. The broad range of clinical syndromes that can develop from the presence of Aspergillus in the respiratory tract is determined by the interaction between host and pathogen. In this review, an oversight of the different clinical entities of pulmonary aspergillosis is given, categorized by their main pathophysiological mechanisms. The underlying immune processes are discussed, and the main clinical, radiological, biochemical, microbiological, and histopathological findings are summarized.
Collapse
Affiliation(s)
- Iris Janssens
- Department of Internal Medicine and Paediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
- VIB Center for Inflammation Research, Ghent, Belgium
| | - Bart N. Lambrecht
- Department of Internal Medicine and Paediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Pulmonary Medicine, ErasmusMC; Rotterdam, The Netherlands
| | - Eva Van Braeckel
- Department of Internal Medicine and Paediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
40
|
Jiang L, Huang Y, Fang M, Chen X, Feng D, Liu J, Jiang Q, Tao R. Dynamic changes of Th1/Th2/Th17 cytokines and hBD-2/3 in erosive oral lichen planus patients saliva before and after prednisone acetate treatment. Heliyon 2024; 10:e24043. [PMID: 38283247 PMCID: PMC10818186 DOI: 10.1016/j.heliyon.2024.e24043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 12/24/2023] [Accepted: 01/02/2024] [Indexed: 01/30/2024] Open
Abstract
Objective This study aimed to investigate the expression of T helper 1 (Th1)/Th2/Th17- related cytokines and human beta defensins 2 and 3 (hBD-2 and -3) in the saliva of patients with erosive oral lichen planus (EOLP) and to explore their role in the pathogenesis of EOLP and the effects of glucocorticoids on EOLP. Methods A total of 30 patients with EOLP and 20 age- and sex-matched healthy individuals were included in this study. The patients were treated with prednisone at a dose of 0.4 mg/(kg·d) for 1 week and examined before and after treatment. Unstimulated whole saliva samples were collected to determine the levels of cytokines (interleukin 1 beta [IL-1β], tumour necrosis factor alpha [TNF]-α, interferon gamma [IFN-γ], IL-4, IL-6, IL-10 and IL-17) by cytometric bead array and those of hBD-2 and -3 b y enzyme-linked immunosorbent assay. In addition, oral rinse samples were collected to detect Candida load. Results The levels of salivary IL-1β, IL-6, hBD-2 and hBD-3 were higher and the IFN-γ/IL-4 and IL-1β/IL-6 ratios were lower in patients with EOLP than in healthy individuals. In patients with EOLP, hBD-2 levels were positively correlated with IFN-γ levels and negatively correlated with IL-17 levels, whereas hBD-3 levels were negatively correlated with IL-17 and IL-10 levels. In addition, the prevalence of EOLP was positively correlated with IL-6 levels and negatively correlated with the IFN-γ/IL-4 ratio. The levels of IL-1β, TNF-α, IFN-γ, IL-6, hBD-2 and hBD-3 and the IFN-γ/IL-4 ratio decreased after treatment with prednisone for 1 week. The levels of IL-6, hBD-2 and hBD-3 were significantly higher in EOLP patients than in healthy individuals; while TNF-α levels and the IFN-γ/IL-4 ratio were significantly lower in EOLP patients than in healthy individuals. Furthermore, the oral counts of Candida spp. (colony forming unit [CFU]) were negatively correlated with TNF-α levels. Numerical Rating Scale(NRS) and Sign scores decreased in EOLP patients after treatment. Approximately 80 % of patients were effectively treated. Salivary TNF-α levels were significantly higher in the treatment-ineffective group than in the treatment-effective group before treatment with prednisone, and differences in salivary IL-6 levels before and after treatment were significantly higher in the treatment-effective group than in the treatment-ineffective group. Conclusions High expression of IL-1β, IL-6, hBD-2 and Th1/Th2 imbalance in saliva may be associated with the pathogenesis of EOLP. IFN-γ/IL-4 balance may serve as a protective factor for EOLP. Glucocorticoids significantly alleviate the symptoms of EOLP and inhibit the expression of Th1/Th2 cytokines.
Collapse
Affiliation(s)
- Lanlan Jiang
- Department of Periodontics and Oral Medicine, College & Hospital of Stomatology, Guangxi Medical University, Guangxi, China
- Guangxi Health Commission Key Laboratory of Prevention and Treatment for Oral Infectious Diseases, Guangxi, China
- Guangxi Key Laboratory of the Rehabilitation and Reconstruction for Oral and Maxillofacial Research, Guangxi, China
| | - Yuxiao Huang
- Department of Periodontics and Oral Medicine, College & Hospital of Stomatology, Guangxi Medical University, Guangxi, China
| | - Meifei Fang
- Department of Periodontics and Oral Medicine, College & Hospital of Stomatology, Guangxi Medical University, Guangxi, China
| | - Xinyu Chen
- Department of Periodontics and Oral Medicine, College & Hospital of Stomatology, Guangxi Medical University, Guangxi, China
| | - Doudou Feng
- Department of Periodontics and Oral Medicine, College & Hospital of Stomatology, Guangxi Medical University, Guangxi, China
| | - Jiaxuan Liu
- Department of Periodontics and Oral Medicine, College & Hospital of Stomatology, Guangxi Medical University, Guangxi, China
| | - Qiaozhi Jiang
- Department of Periodontics and Oral Medicine, College & Hospital of Stomatology, Guangxi Medical University, Guangxi, China
| | - Renchuan Tao
- Department of Periodontics and Oral Medicine, College & Hospital of Stomatology, Guangxi Medical University, Guangxi, China
- Guangxi Health Commission Key Laboratory of Prevention and Treatment for Oral Infectious Diseases, Guangxi, China
- Guangxi Key Laboratory of the Rehabilitation and Reconstruction for Oral and Maxillofacial Research, Guangxi, China
| |
Collapse
|
41
|
Höft MA, Duvenage L, Salie S, Keeton R, Botha A, Schwartz IS, Govender NP, Brown GD, Hoving JC. The pathogenesis of experimental Emergomycosis in mice. PLoS Negl Trop Dis 2024; 18:e0011850. [PMID: 38198478 PMCID: PMC10805315 DOI: 10.1371/journal.pntd.0011850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 01/23/2024] [Accepted: 12/07/2023] [Indexed: 01/12/2024] Open
Abstract
Emergomyces africanus is a recently identified thermally-dimorphic fungal pathogen that causes disseminated infection in people living with advanced HIV disease. Known as emergomycosis, this disseminated disease is associated with very high case fatality rates. Over the last decade, improved diagnostics and fungal identification in South Africa resulted in a dramatic increase in the number of reported cases. Although the true burden of disease is still unknown, emergomycosis is among the most frequently diagnosed dimorphic fungal infections in Southern Africa; and additional species in the genus have been identified on four continents. Little is known about the pathogenesis and the host's immune response to this emerging pathogen. Therefore, we established a murine model of pulmonary infection using a clinical isolate, E. africanus (CBS 136260). Both conidia and yeast forms caused pulmonary and disseminated infection in mice with organisms isolated in culture from lung, spleen, liver, and kidney. Wild-type C57BL/6 mice demonstrated a drop in body weight at two weeks post-infection, corresponding to a peak in fungal burden in the lung, spleen, liver, and kidney. An increase in pro-inflammatory cytokine production was detected in homogenized lung supernatants including IFN-γ, IL-1β, IL-6, IL12-p40 and IL-17 at three- and four-weeks post-infection. No significant differences in TNF, IL-12p70 and IL-10 were observed in wild-type mice between one and four-weeks post-infection. Rag-1-deficient mice, lacking mature T-and B-cells, had an increased fungal burden associated with reduced IFN-γ production. Together our data support a protective T-helper type-1 immune response to E. africanus infection. This may provide a possible explanation for the susceptibility of only a subset of people living with advanced HIV disease despite hypothesized widespread environmental exposure. In summary, we have established a novel murine model of E. africanus disease providing critical insights into the host immune components required for eliminating the infection.
Collapse
Affiliation(s)
- Maxine A. Höft
- CMM AFRICA Medical Mycology Research Unit, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Lucian Duvenage
- CMM AFRICA Medical Mycology Research Unit, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sumayah Salie
- CMM AFRICA Medical Mycology Research Unit, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Roanne Keeton
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Alfred Botha
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa
| | - Ilan S. Schwartz
- Division of Infectious Diseases, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Nelesh P. Govender
- CMM AFRICA Medical Mycology Research Unit, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- National Institute for Communicable Diseases, Division of the National Health Laboratory Service, Johannesburg, South Africa; School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Medical Research Council Centre for Medical Mycology at the University of Exeter, Geoffrey Pope Building Stocker Road, Exeter, United Kingdom
| | - Gordon D. Brown
- CMM AFRICA Medical Mycology Research Unit, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- Medical Research Council Centre for Medical Mycology at the University of Exeter, Geoffrey Pope Building Stocker Road, Exeter, United Kingdom
| | - Jennifer Claire Hoving
- CMM AFRICA Medical Mycology Research Unit, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Medical Research Council Centre for Medical Mycology at the University of Exeter, Geoffrey Pope Building Stocker Road, Exeter, United Kingdom
| |
Collapse
|
42
|
Petrucelli MF, Martins-Santana L, Sanches PR, Oliveira VM, Rossi A, Martinez-Rossi NM. The Transcription Factor StuA Regulates the Glyoxylate Cycle in the Dermatophyte Trichophyton rubrum under Carbon Starvation. Int J Mol Sci 2023; 25:405. [PMID: 38203573 PMCID: PMC10778625 DOI: 10.3390/ijms25010405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024] Open
Abstract
Trichophyton rubrum is the primary causative agent of dermatophytosis worldwide. This fungus colonizes keratinized tissues and uses keratin as a nutritional source during infection. In T. rubrum-host interactions, sensing a hostile environment triggers the adaptation of its metabolic machinery to ensure its survival. The glyoxylate cycle has emerged as an alternative metabolic pathway when glucose availability is limited; this enables the conversion of simple carbon compounds into glucose via gluconeogenesis. In this study, we investigated the impact of stuA deletion on the response of glyoxylate cycle enzymes during fungal growth under varying culture conditions in conjunction with post-transcriptional regulation through alternative splicing of the genes encoding these enzymes. We revealed that the ΔstuA mutant downregulated the malate synthase and isocitrate lyase genes in a keratin-containing medium or when co-cultured with human keratinocytes. Alternative splicing of an isocitrate lyase gene yielded a new isoform. Enzymatic activity assays showed specific instances where isocitrate lyase and malate synthase activities were affected in the mutant strain compared to the wild type strain. Taken together, our results indicate a relevant balance in transcriptional regulation that has distinct effects on the enzymatic activities of malate synthase and isocitrate lyase.
Collapse
Affiliation(s)
| | | | | | | | | | - Nilce M. Martinez-Rossi
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil; (M.F.P.); (L.M.-S.); (P.R.S.); (V.M.O.); (A.R.)
| |
Collapse
|
43
|
Özdirik B, Schnabl B. Microbial Players in Primary Sclerosing Cholangitis: Current Evidence and Concepts. Cell Mol Gastroenterol Hepatol 2023; 17:423-438. [PMID: 38109970 PMCID: PMC10837305 DOI: 10.1016/j.jcmgh.2023.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/12/2023] [Accepted: 12/12/2023] [Indexed: 12/20/2023]
Abstract
Primary sclerosing cholangitis (PSC) is a rare cholestatic liver disease with progressive biliary inflammation, destruction of the biliary tract, and fibrosis, resulting in liver cirrhosis and end-stage liver disease. To date, liver transplantation is the only definitive treatment option for PSC. The precise etiology of PSC remains elusive, but it is widely accepted to involve a complex interplay between genetic predisposition, immunologic dysfunction, and environmental influence. In recent years, the gut-liver axis has emerged as a crucial pathway contributing to the pathogenesis of PSC, with particular focus on the role of gut microbiota. However, the role of the fungal microbiome or mycobiome has been overlooked for years, resulting in a lack of comprehensive studies on its involvement in PSC. In this review, we clarify the present clinical and mechanistic data and concepts concerning the gut bacterial and fungal microbiota in the context of PSC. This review sheds light on the role of specific microbes and elucidates the dynamics of bacterial and fungal populations. Moreover, we discuss the latest insights into microbe-altering therapeutic approaches involving the gut-liver axis and bile acid metabolism.
Collapse
Affiliation(s)
- Burcin Özdirik
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, California; Department of Medicine, VA San Diego Healthcare System, San Diego, California.
| |
Collapse
|
44
|
Wang G, Li Q, Guo Y, Chen L, Yao Y, Zhong Y, Sun J, Yan X, Wang H, Wang X, Ding L, Ju H. Interception Proximity Labeling for Interrogating Cell Efflux Microenvironment. Anal Chem 2023; 95:17798-17807. [PMID: 37976298 DOI: 10.1021/acs.analchem.3c03879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
The difficulty in elucidating the microenvironment of extracellular H2O2 efflux has led to the lack of a critical extracellular link in studies of the mechanisms of redox signaling pathways. Herein, we mounted horseradish peroxidase (HRP) to glycans expressed globally on the living cell surface and constructed an interception proximity labeling (IPL) platform for H2O2 efflux. The release of endogenous H2O2 is used as a "physiological switch" for HRP to enable proximity labeling. Using this platform, we visualize the oxidative stress state of tumor cells under the condition of nutrient withdrawal, as well as that of macrophages exposed to nonparticulate stimuli. Furthermore, in combination with a proteomics technique, we identify candidate proteins at the invasion interface between fungal mimics (zymosan) and macrophages by interception labeling of locally accumulated H2O2 and confirm that Toll-like receptor 2 binds zymosan in a glycan-dependent manner. The IPL platform has great potential to elucidate the mechanisms underlying biological processes involving redox pathways.
Collapse
Affiliation(s)
- Guyu Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Qiang Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yuna Guo
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Liusheng Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yunyan Yao
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yihong Zhong
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Jiahui Sun
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Xiaomin Yan
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Hongwei Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China
| | - Xiaojian Wang
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Lin Ding
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|
45
|
Earle K, Valero C, Conn DP, Vere G, Cook PC, Bromley MJ, Bowyer P, Gago S. Pathogenicity and virulence of Aspergillus fumigatus. Virulence 2023; 14:2172264. [PMID: 36752587 PMCID: PMC10732619 DOI: 10.1080/21505594.2023.2172264] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 12/16/2022] [Indexed: 02/09/2023] Open
Abstract
Pulmonary infections caused by the mould pathogen Aspergillus fumigatus are a major cause of morbidity and mortality globally. Compromised lung defences arising from immunosuppression, chronic respiratory conditions or more recently, concomitant viral or bacterial pulmonary infections are recognised risks factors for the development of pulmonary aspergillosis. In this review, we will summarise our current knowledge of the mechanistic basis of pulmonary aspergillosis with a focus on emerging at-risk populations.
Collapse
Affiliation(s)
- Kayleigh Earle
- Manchester Fungal Infection Group, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Clara Valero
- Manchester Fungal Infection Group, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Daniel P. Conn
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - George Vere
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Peter C. Cook
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Michael J. Bromley
- Manchester Fungal Infection Group, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Paul Bowyer
- Manchester Fungal Infection Group, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Sara Gago
- Manchester Fungal Infection Group, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| |
Collapse
|
46
|
Fernandes C, Casadevall A, Gonçalves T. Mechanisms of Alternaria pathogenesis in animals and plants. FEMS Microbiol Rev 2023; 47:fuad061. [PMID: 37884396 DOI: 10.1093/femsre/fuad061] [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: 05/08/2023] [Revised: 09/18/2023] [Accepted: 10/25/2023] [Indexed: 10/28/2023] Open
Abstract
Alternaria species are cosmopolitan fungi darkly pigmented by melanin that infect numerous plant species causing economically important agricultural spoilage of various food crops. Alternaria spp. also infect animals, being described as entomopathogenic fungi but also infecting warm-blooded animals, including humans. Their clinical importance in human health, as infection agents, lay in the growing number of immunocompromised patients. Moreover, Alternaria spp. are considered some of the most abundant and potent sources of airborne sensitizer allergens causing allergic respiratory diseases, as severe asthma. Among the numerous strategies deployed by Alternaria spp. to attack their hosts, the production of toxins, carrying critical concerns to public health as food contaminant, and the production of hydrolytic enzymes such as proteases, can be highlighted. Alternaria proteases also trigger allergic symptoms in individuals with fungal sensitization, acting as allergens and facilitating antigen access to the host subepithelium. Here, we review the current knowledge about the mechanisms of Alternaria pathogenesis in plants and animals, the strategies used by Alternaria to cope with the host defenses, and the involvement Alternaria allergens and mechanisms of sensitization.
Collapse
Affiliation(s)
- Chantal Fernandes
- CNC-UC - Center for Neuroscience and Cell Biology of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Wolfe Street, Room E5132, Baltimore, Maryland 21205, USA
| | - Teresa Gonçalves
- CNC-UC - Center for Neuroscience and Cell Biology of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal
- FMUC - Faculty of Medicine, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal
| |
Collapse
|
47
|
Daud M, Dasari P, Adelfinger M, Langenhorst D, Lother J, Slavkovic-Lukic D, Berges C, Kruhm M, Galler A, Schleussner C, Luther CH, Alberter K, Althammer A, Shaikh H, Pallmann N, Bodem J, El-Mowafy M, Beilhack A, Dittrich M, Topp MS, Zipfel PF, Beyersdorf N. Enolase 1 of Candida albicans binds human CD4 + T cells and modulates naïve and memory responses. Eur J Immunol 2023; 53:e2250284. [PMID: 37503840 DOI: 10.1002/eji.202250284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 06/05/2023] [Accepted: 07/27/2023] [Indexed: 07/29/2023]
Abstract
To obtain a better understanding of the biology behind life-threatening fungal infections caused by Candida albicans, we recently conducted an in silico screening for fungal and host protein interaction partners. We report here that the extracellular domain of human CD4 binds to the moonlighting protein enolase 1 (Eno1) of C. albicans as predicted bioinformatically. By using different anti-CD4 monoclonal antibodies, we determined that C. albicans Eno1 (CaEno1) primarily binds to the extracellular domain 3 of CD4. Functionally, we observed that CaEno1 binding to CD4 activated lymphocyte-specific protein tyrosine kinase (LCK), which was also the case for anti-CD4 monoclonal antibodies tested in parallel. CaEno1 binding to naïve human CD4+ T cells skewed cytokine secretion toward a Th2 profile indicative of poor fungal control. Moreover, CaEno1 inhibited human memory CD4+ T-cell recall responses. Therapeutically, CD4+ T cells transduced with a p41/Crf1-specific T-cell receptor developed for adoptive T-cell therapy were not inhibited by CaEno1 in vitro. Together, the interaction of human CD4+ T cells with CaEno1 modulated host CD4+ T-cell responses in favor of the fungus. Thus, CaEno1 mediates not only immune evasion through its interference with complement regulators but also through the direct modulation of CD4+ T-cell responses.
Collapse
Affiliation(s)
- Muhammad Daud
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Prasad Dasari
- Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany
| | - Marion Adelfinger
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | - Daniela Langenhorst
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Jasmin Lother
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | - Dragana Slavkovic-Lukic
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | - Carsten Berges
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | - Michaela Kruhm
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | | | | | | | - Karl Alberter
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Anton Althammer
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Haroon Shaikh
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | - Niklas Pallmann
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Jochen Bodem
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Mohammed El-Mowafy
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
- Faculty of Pharmacy, Department of Microbiology & Immunology, Mansoura University, Mansoura, Egypt
| | - Andreas Beilhack
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | - Marcus Dittrich
- Chair of Bioinformatics, University of Würzburg, Würzburg, Germany
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Max S Topp
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | - Peter F Zipfel
- Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany
- Friedrich Schiller University, Jena, Germany
| | - Niklas Beyersdorf
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
48
|
Munyemana MA, Pande A, Kallogjeri D, Farrell NF, Schneider JS, Kendall PL, Roland LT. Invasive fungal sinusitis risk factors among immunosuppressed hematopoietic stem cell transplant recipients. Int Forum Allergy Rhinol 2023; 13:2113-2118. [PMID: 37260282 PMCID: PMC10615690 DOI: 10.1002/alr.23200] [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: 04/06/2023] [Revised: 05/19/2023] [Accepted: 05/19/2023] [Indexed: 06/02/2023]
Abstract
KEY POINTS Invasive fungal sinusitis (IFS) rate and risk factors in transplant recipients were explored IFS rate is higher in allogeneic recipients with prior transplants and worse comorbidity scores The at-risk timeframes for IFS development were identified.
Collapse
Affiliation(s)
- Marie-Ange Munyemana
- Clinical Outcomes Research Office, Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine in St Louis, St Louis, Missouri, USA
- Cooper Medical School of Rowan University, Camden, New Jersey, USA
| | - Anupam Pande
- Department of Medicine, Division of Infectious Disease, Washington University School of Medicine in St Louis, St Louis, Missouri, USA
| | - Dorina Kallogjeri
- Clinical Outcomes Research Office, Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine in St Louis, St Louis, Missouri, USA
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine in St Louis, St Louis, Missouri, USA
| | - Nyssa F Farrell
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine in St Louis, St Louis, Missouri, USA
| | - John S Schneider
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine in St Louis, St Louis, Missouri, USA
| | - Peggy L Kendall
- Department of Medicine, Division of Allergy and Immunology, Washington University School of Medicine in St Louis, St Louis, Missouri, USA
| | - Lauren T Roland
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine in St Louis, St Louis, Missouri, USA
| |
Collapse
|
49
|
Liu Q, Zhang W, Pei Y, Tao H, Ma J, Li R, Zhang F, Wang L, Shen L, Liu Y, Jia X, Hu Y. Gut mycobiome as a potential non-invasive tool in early detection of lung adenocarcinoma: a cross-sectional study. BMC Med 2023; 21:409. [PMID: 37904139 PMCID: PMC10617124 DOI: 10.1186/s12916-023-03095-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/26/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND The gut mycobiome of patients with lung adenocarcinoma (LUAD) remains unexplored. This study aimed to characterize the gut mycobiome in patients with LUAD and evaluate the potential of gut fungi as non-invasive biomarkers for early diagnosis. METHODS In total, 299 fecal samples from Beijing, Suzhou, and Hainan were collected prospectively. Using internal transcribed spacer 2 sequencing, we profiled the gut mycobiome. Five supervised machine learning algorithms were trained on fungal signatures to build an optimized prediction model for LUAD in a discovery cohort comprising 105 patients with LUAD and 61 healthy controls (HCs) from Beijing. Validation cohorts from Beijing, Suzhou, and Hainan comprising 44, 17, and 15 patients with LUAD and 26, 19, and 12 HCs, respectively, were used to evaluate efficacy. RESULTS Fungal biodiversity and richness increased in patients with LUAD. At the phylum level, the abundance of Ascomycota decreased, while that of Basidiomycota increased in patients with LUAD. Candida and Saccharomyces were the dominant genera, with a reduction in Candida and an increase in Saccharomyces, Aspergillus, and Apiotrichum in patients with LUAD. Nineteen operational taxonomic unit markers were selected, and excellent performance in predicting LUAD was achieved (area under the curve (AUC) = 0.9350) using a random forest model with outcomes superior to those of four other algorithms. The AUCs of the Beijing, Suzhou, and Hainan validation cohorts were 0.9538, 0.9628, and 0.8833, respectively. CONCLUSIONS For the first time, the gut fungal profiles of patients with LUAD were shown to represent potential non-invasive biomarkers for early-stage diagnosis.
Collapse
Affiliation(s)
- Qingyan Liu
- Graduate School, Chinese People's Liberation Army Medical School, Beijing, China
- Department of Oncology, Fifth Medical Center of the Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Haidian Distrist, Beijing, 100000, China
| | - Weidong Zhang
- Graduate School, Chinese People's Liberation Army Medical School, Beijing, China
- Department of Thoracic Surgery, First Medical Center of the Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100000, China
| | - Yanbin Pei
- Graduate School, Chinese People's Liberation Army Medical School, Beijing, China
| | - Haitao Tao
- Department of Oncology, Fifth Medical Center of the Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Haidian Distrist, Beijing, 100000, China
| | - Junxun Ma
- Department of Oncology, Fifth Medical Center of the Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Haidian Distrist, Beijing, 100000, China
| | - Rong Li
- Department of Health Medicine, Second Medical Center of the Chinese People's Liberation Army General Hospital, Beijing, China
| | - Fan Zhang
- Department of Oncology, Fifth Medical Center of the Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Haidian Distrist, Beijing, 100000, China
| | - Lijie Wang
- Department of Oncology, Fifth Medical Center of the Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Haidian Distrist, Beijing, 100000, China
| | - Leilei Shen
- Department of Thoracic Surgery, Hainan Medical Center of the Chinese People's Liberation Army General Hospital, Hainan, China
| | - Yang Liu
- Department of Thoracic Surgery, First Medical Center of the Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100000, China.
| | - Xiaodong Jia
- Department of Oncology, Fifth Medical Center of the Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Haidian Distrist, Beijing, 100000, China.
| | - Yi Hu
- Department of Oncology, Fifth Medical Center of the Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Haidian Distrist, Beijing, 100000, China.
| |
Collapse
|
50
|
Rashidi M, Bazi A, Ahmadzadeh A, Romeo O, Rezaei-Matehkolaei A, Abastabar M, Haghani I, Mirzaei S. The growth inhibitory and apoptotic effects of umbelliprenin in a mouse model of systemic candidiasis. J Appl Microbiol 2023; 134:lxad201. [PMID: 37669891 DOI: 10.1093/jambio/lxad201] [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: 06/12/2023] [Revised: 08/23/2023] [Accepted: 09/02/2023] [Indexed: 09/07/2023]
Abstract
AIMS Umbelliprenin has shown promising biological activities, including immunoregulatory, anti-inflammatory, and anti-cancer effects. The present study investigated the growth inhibitory and apoptotic effects of umbelliprenin against Candida albicans in a BALB/c mice model of disseminated candidiasis. METHODS AND RESULTS First, an antimicrobial assay via microdilution sensitivity test was performed. Then, twenty-five 6-week-old female BALB/c mice (20 ± 12 g) were divided into five groups of five mice, including one control group (no umbelliprenin treatment) and four experimental groups: C. albicans-infected mice treated with umbelliprenin at the doses of 5, 10, 20, and 40 mg kg -1. The brain, lung, kidney, spleen, and liver tissues were examined for fungal infection and histological lesions, and TUNEL staining was performed to assess apoptosis. The β-1, 3-glucan synthase assay was used to evaluate enzymatic activity, and gene expression analysis was also performed to investigate the transcriptional changes of ERG11, CDR1, ALS1, and HWP1 genes. The MIC of umbelliprenin was 1.5 mg mL-1. Our results showed that at the 40 mg kg -1 dose, umbelliprenin was able to eradicate fungal infection in BALB/c mice. The percentage of apoptotic cells in umbelliprenin-treated groups increased in a concentration-dependent manner. Umbelliprenin (40 mg kg -1) also inhibited the expression of β-1, 3-glucan synthase, and the genes involved in antifungal resistance (CDR1 and ERG11), as well as the expression of the genes encoding adhesins (ALS1 and HWP1). CONCLUSION Our results showed that umbelliprenin could promote antifungal effects, partly via inducing apoptosis.
Collapse
Affiliation(s)
- Mohsen Rashidi
- Department of Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari 48157-33971, Iran
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari 48157-33971, Iran
| | - Ali Bazi
- Department of Laboratory Sciences, School of Allied Medical Sciences, Zabol University of Medical Sciences, Zabol 98616-15881, Iran
| | - Alireza Ahmadzadeh
- Department of Lab Sciences, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 19839-69411, Iran
| | - Orazio Romeo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 ME, Italy
- IRCCS-Centro Neurolesi Bonino-Pulejo, 98124 ME, Italy
| | - Ali Rezaei-Matehkolaei
- Department of Medical Mycology, School of Medicine, Infectious and Tropical Diseases Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 61357-15794, Iran
| | - Mahdi Abastabar
- Invasive Fungi Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari 48157-33971, Iran
- Department of Medical Mycology, School of Medicine, Mazandaran University of Medical Sciences, Sari 48157-33971, Iran
| | - Iman Haghani
- Invasive Fungi Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari 48157-33971, Iran
- Department of Medical Mycology, School of Medicine, Mazandaran University of Medical Sciences, Sari 48157-33971, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Sciences, Islamic Azad University Science and Research Branch, Tehran 14778-93855, Iran
| |
Collapse
|