1
|
Li X, Wang Q, Huang J, Yue X, Zhang X, Fan X, Fang Z, Wang G, Qiu Z, Luo D, Guo Q, Zhuang AX, Zhan S, Li Q, Zhao Z. Posaconazole nanocrystals dry powder inhalers for the local treatment of invasive pulmonary aspergillosis. Int J Pharm 2025; 668:124938. [PMID: 39557177 DOI: 10.1016/j.ijpharm.2024.124938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024]
Abstract
Invasive pulmonary aspergillosis poses a significant threat to immunocompromised patients, characterized by high mortality rates. Posaconazole (PSZ), a second-generation triazole antifungal, exhibits broad-spectrum activity but suffers from limited pulmonary concentrations and notable systemic side effects when administered orally or intravenously. This study focuses on optimizing PSZ nanocrystals-agglomerated particles for dry powder inhalers (DPIs) to enhance solubility, dissolution rates, and pulmonary deposition, ultimately improving therapeutic efficacy while minimizing systemic adverse effects. We employed wet medium milling and spray-drying techniques to formulate PSZ nanocrystals-agglomerated DPIs. Various stabilizers including HPMC, HPC, Soluplus, and PVPK30, were systematically evaluated to optimize physicochemical properties. Aerosolization performance was assessed using the Next Generation Impactor, while antifungal efficacy was evaluated through in vitro and in vivo studies. The optimized PSZ DPIs demonstrated significant enhancements in solubility and dissolution rates, with a fine particle fraction (FPF) of 78.58 ± 3.21%, ensuring optimal lung delivery. In vitro experiments revealed potent effects with minimal cytotoxicity to lung cells. In vivo studies indicated that the optimized formulation achieved a Cmax/AUC0→∞ ratio in lung tissues that was 27.32 and 6.76-fold higher than that of the oral suspension, highlighting increased local drug concentrations. This approach presents a scalable, cost-effective strategy for the pulmonary delivery of PSZ, ensuring high drug loading and promising clinical outcomes in treating pulmonary fungal infections.
Collapse
Affiliation(s)
- Xuchun Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Qing Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jiewen Huang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xiao Yue
- College of Pharmacy, Jinan University, Guangzhou 510006, Guangdong, China
| | - Xuejuan Zhang
- College of Pharmacy, Jinan University, Guangzhou 510006, Guangdong, China
| | - Xinxin Fan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Zhian Fang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Guanlin Wang
- College of Pharmacy, Jinan University, Guangzhou 510006, Guangdong, China
| | - Zhenwen Qiu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Dandong Luo
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Qiupin Guo
- Drug Non-Clinical Evaluation and Research Center of Guangzhou General Pharmaceutical Research Institute, Guangzhou 510240, China
| | - Alan Xiaodong Zhuang
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, UK.
| | - Shaofeng Zhan
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Qingguo Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Ziyu Zhao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510006, China.
| |
Collapse
|
2
|
Tesfamariam M, Vij R, Trümper V, Hube B, Brunke S. Shining a light on Candida-induced epithelial damage with a luciferase reporter. mSphere 2024; 9:e0050924. [PMID: 39412273 PMCID: PMC11580449 DOI: 10.1128/msphere.00509-24] [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: 06/13/2024] [Accepted: 09/17/2024] [Indexed: 11/22/2024] Open
Abstract
Host cell damage is a key parameter for research in infection biology, drug testing, and substance safety screening. In this study, we introduce a luciferase reporter system as a new and reliable assay to measure cell damage and validate it with the pathogenic yeast, Candida albicans, as a test case. We transduced human epithelial cell lines with a lentiviral vector to stably express an optimized luciferase enzyme, Nanoluc. Upon cell damage, the release of cytoplasmic luciferase into the extracellular space can be easily detected by a luminometer. We used the luciferase reporter system to investigate the damage caused by C. albicans to different newly generated epithelial reporter cell lines. We found that fungus-induced cell damage, as determined by established methods, correlated tightly with the release of the luciferase. The new luciferase reporter system is a simple, sensitive, robust, and inexpensive method for measuring host cell damage and has a sensitivity comparable to the standard assay, release of lactate dehydrogenase. It is suitable for high-throughput studies of pathogenesis mechanisms of any microbe, for antimicrobial drug screening, and many other applications.IMPORTANCEWe present a quick, easy, inexpensive, and reliable assay to measure damage to mammalian cells. To this end, we created reporter cell lines which artificially express luciferase, an enzyme that can be easily detected in the supernatant when these cells are damaged. We used infections with the well-investigated fungal pathogen of humans, Candida albicans, as a test case of our system. Using our reporter, we were able to recapitulate the known effects of strain variability, gene deletions, and antifungal treatments on host cell damage. This easily adaptable reporter system can be used to screen for damage in infection models with different microbial species, assay cell-damaging potential of substances, discover new non-toxic antibiotics, and many other damage-based applications.
Collapse
Affiliation(s)
- Millen Tesfamariam
- Department of Molecular Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knoell Institute, Jena, Germany
| | - Raghav Vij
- Department of Molecular Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knoell Institute, Jena, Germany
| | - Verena Trümper
- Department of Molecular Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knoell Institute, Jena, Germany
| | - Bernhard Hube
- Department of Molecular Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knoell Institute, Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
| | - Sascha Brunke
- Department of Molecular Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knoell Institute, Jena, Germany
| |
Collapse
|
3
|
Grajales-Urrego DM, Mantilla-Sylvain F, Rolon-Cadena MC, Basto-Borbón WM, Álvarez-Figueroa J. Venous intestinal ischemia of fungal origin as a cause of intestinal obstruction in immunocompromised patients: case report and literature review. Gut Pathog 2024; 16:66. [PMID: 39523338 PMCID: PMC11552342 DOI: 10.1186/s13099-024-00658-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Mucormycosis is a highly lethal opportunistic fungal disease caused by ubiquitous molds of the order Mucorales, with Rhizopus, Lichtheimia and Mucor being the most common genera. This rare disease primarily affects immunocompromised patients, with presentations ranging from rhino-orbito-cerebral infections to disseminated mucormycosis with angioinvasion, leading to thrombosis and tissue infarction. Gastrointestinal mucormycosis is the least common clinical presentation and is believed to be secondary to spore ingestion. It can involve multiple components of the gastrointestinal tract, such as the stomach, liver, ileum, and colon, with nonspecific manifestations, including pain, nausea, vomiting, and abdominal distension. The initial clinical presentation may even manifest as gastrointestinal bleeding due to gastric ulceration or intestinal perforation. CASE PRESENTATION Here we present the case of a 48-year-old male patient with a 9-year history of human immunodeficiency virus (HIV) infection who was hospitalized in the context of febrile neutropenia and whose acute respiratory infection was documented; therefore, antibiotic treatment was initiated. However, due to persistent febrile peaks and peripheral blood showing documentation of multilineage cytopenias, a bone marrow biopsy was performed, compatible with presenting features of marrow myelodysplasia. During hospitalization, the patient presented left flank abdominal pain, and an abdominal computed tomography (CT) scan revealed signs of intussusception of a small bowel loop at the distal jejunum level, leading to intestinal obstruction with ischemic progression, requiring ileectomy (60 cm). Histopathological analysis of the resected intestine revealed severe transmural ischemic changes associated with venous thrombosis due to fungal structures, with histochemical studies demonstrating the presence of zygomycete (Mucor) fungal structures, leading to the initiation of treatment with amphotericin B. However, despite treatment, the patient experienced progressive clinical deterioration with persistent fever and ventilatory failure, with follow-up tests showing absolute neutropenia and blood cultures positive for yeast, leading to death 52 days after admission. CONCLUSIONS The diagnosis of intestinal mucormycosis may be delayed due to the lack of specificity of the signs and symptoms. Pathologists as well as histopathological studies are essential for timely treatment.
Collapse
Affiliation(s)
| | | | | | | | - Johanna Álvarez-Figueroa
- Department of Pathology and Laboratories, Fundación Santa Fe de Bogotá, Bogotá, D.C, Colombia.
- School of Medicine, Universidad de los Andes, Bogotá, D.C, Colombia.
| |
Collapse
|
4
|
Brown GD, Ballou ER, Bates S, Bignell EM, Borman AM, Brand AC, Brown AJP, Coelho C, Cook PC, Farrer RA, Govender NP, Gow NAR, Hope W, Hoving JC, Dangarembizi R, Harrison TS, Johnson EM, Mukaremera L, Ramsdale M, Thornton CR, Usher J, Warris A, Wilson D. The pathobiology of human fungal infections. Nat Rev Microbiol 2024; 22:687-704. [PMID: 38918447 DOI: 10.1038/s41579-024-01062-w] [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] [Accepted: 05/14/2024] [Indexed: 06/27/2024]
Abstract
Human fungal infections are a historically neglected area of disease research, yet they cause more than 1.5 million deaths every year. Our understanding of the pathophysiology of these infections has increased considerably over the past decade, through major insights into both the host and pathogen factors that contribute to the phenotype and severity of these diseases. Recent studies are revealing multiple mechanisms by which fungi modify and manipulate the host, escape immune surveillance and generate complex comorbidities. Although the emergence of fungal strains that are less susceptible to antifungal drugs or that rapidly evolve drug resistance is posing new threats, greater understanding of immune mechanisms and host susceptibility factors is beginning to offer novel immunotherapeutic options for the future. In this Review, we provide a broad and comprehensive overview of the pathobiology of human fungal infections, focusing specifically on pathogens that can cause invasive life-threatening infections, highlighting recent discoveries from the pathogen, host and clinical perspectives. We conclude by discussing key future challenges including antifungal drug resistance, the emergence of new pathogens and new developments in modern medicine that are promoting susceptibility to infection.
Collapse
Affiliation(s)
- Gordon D Brown
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK.
| | - Elizabeth R Ballou
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Steven Bates
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Elaine M Bignell
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Andrew M Borman
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Alexandra C Brand
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Alistair J P Brown
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Carolina Coelho
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Peter C Cook
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Rhys A Farrer
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Nelesh P Govender
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Neil A R Gow
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - William Hope
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - J Claire Hoving
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Rachael Dangarembizi
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Thomas S Harrison
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Elizabeth M Johnson
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Liliane Mukaremera
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Mark Ramsdale
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | | | - Jane Usher
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Adilia Warris
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Duncan Wilson
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| |
Collapse
|
5
|
Ali B, Chauhan A, Kumar M, Kumar P, Carolus H, Lobo Romero C, Vergauwen R, Singh A, Banerjee A, Prakash A, Rudramurthy SM, Van Dijck P, Ibrahim AS, Prasad R. A Comprehensive Analysis of the Lipidomic Signatures in Rhizopus delemar. J Fungi (Basel) 2024; 10:760. [PMID: 39590679 PMCID: PMC11595932 DOI: 10.3390/jof10110760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/23/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Certain species of Mucorales have been identified as causative agents of mucormycosis, a rare yet often lethal fungal infection. Notably, these fungi exhibit intrinsic resistance to common azole drugs, which target lipids. Given the pivotal role of lipids in drug resistance and their contribution to innate resistance to azoles, this study provides a comprehensive overview of key lipid classes, including sphingolipids (SLs), glycerophospholipids (GPLs), and sterols, in Rhizopus delemar 99-880, a well-characterized reference strain among Mucorales. Using shotgun lipidomics as well as liquid- and gas-chromatography-based mass spectrometric analyses, we identified the lipid intermediates and elucidated the biosynthetic pathways of SLs, PGLs, and sterols. The acidic SLs were not found, probably because the acidic branch of the SL biosynthesis pathway terminates at α-hydroxy phytoceramides, as evident by their high abundance. Intermediates in the neutral SL pathway incorporated higher levels of 16:0 fatty acid compared to other pathogenic fungi. A strikingly high phosphatidylethanolamine (PE)/phosphatdylcholine (PC) ratio was observed among GPLs. Ergosterol remains the major sterol, similar to other fungi, and our analysis confirms the existence of alternate ergosterol biosynthesis pathways. The total lipidomic profile of R. delemar 99-880 offers insights into its lipid metabolism and potential implications for studying pathogenesis and drug resistance mechanisms.
Collapse
Affiliation(s)
- Basharat Ali
- Amity Institute of Integrative Science and Health, Amity University Gurugram, Gurugram 122413, India
- Amity Institute of Biotechnology, Amity University Gurugram, Gurugram 122413, India
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Anshu Chauhan
- Amity Institute of Integrative Science and Health, Amity University Gurugram, Gurugram 122413, India
- Amity Institute of Biotechnology, Amity University Gurugram, Gurugram 122413, India
| | - Mohit Kumar
- Yeast Biofuel Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Praveen Kumar
- Amity Institute of Integrative Science and Health, Amity University Gurugram, Gurugram 122413, India
- Amity Institute of Biotechnology, Amity University Gurugram, Gurugram 122413, India
| | - Hans Carolus
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Celia Lobo Romero
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Rudy Vergauwen
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Ashutosh Singh
- Department of Biochemistry, University of Lucknow, Lucknow 226007, India
| | - Atanu Banerjee
- Amity Institute of Integrative Science and Health, Amity University Gurugram, Gurugram 122413, India
- Amity Institute of Biotechnology, Amity University Gurugram, Gurugram 122413, India
| | - Amresh Prakash
- Amity Institute of Integrative Science and Health, Amity University Gurugram, Gurugram 122413, India
- Amity Institute of Biotechnology, Amity University Gurugram, Gurugram 122413, India
| | - Shivaprakash M. Rudramurthy
- Department of Medical Microbiology, Postgraduate Institute of Medical Education & Research, Chandigarh 160012, India
| | - Patrick Van Dijck
- Laboratory of Molecular Cell Biology, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Ashraf S. Ibrahim
- Division of Infectious Diseases, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
- David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Rajendra Prasad
- Amity Institute of Integrative Science and Health, Amity University Gurugram, Gurugram 122413, India
- Amity Institute of Biotechnology, Amity University Gurugram, Gurugram 122413, India
| |
Collapse
|
6
|
Kapitan M, Niemiec MJ, Millet N, Brandt P, Chowdhury MEK, Czapka A, Abdissa K, Hoffmann F, Lange A, Veleba M, Nietzsche S, Mosig AS, Löffler B, Marquet M, Makarewicz O, Kline KA, Vylkova S, Swidergall M, Jacobsen ID. Synergistic cross-kingdom host cell damage between Candida albicans and Enterococcus faecalis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612452. [PMID: 39314435 PMCID: PMC11419042 DOI: 10.1101/2024.09.11.612452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
The fungus Candida albicans and the Gram-positive bacterium Enterococcus faecalis share mucosal niches in the human body. As opportunistic pathogens, both are found to expand population size during dysbiosis, and can cause severe systemic infections in susceptible individuals. Here, we show that the presence of C. albicans results in increased host cell damage by E. faecalis . Furthermore, E. faecalis aggravates oropharyngeal candidiasis in mice. Increased damage is mediated by enterococcal cytolysin, and involves both physical interaction and altered glucose availability. Physical interaction promotes accumulation of bacteria on host cells, facilitating contact of cytolysin with host cells. Glucose depletion by the metabolic activity of the fungus sensitized host cells to cytolysin. This work illustrates how a complex interplay between fungi and bacteria can result in detrimental consequences for the host.
Collapse
|
7
|
Kavaliauskas P, Gu Y, Hasin N, Graf KT, Alqarihi A, Shetty AC, McCracken C, Walsh TJ, Ibrahim AS, Bruno VM. Multiple roles for hypoxia inducible factor 1-alpha in airway epithelial cells during mucormycosis. Nat Commun 2024; 15:5282. [PMID: 38902255 PMCID: PMC11190229 DOI: 10.1038/s41467-024-49637-8] [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: 06/21/2023] [Accepted: 06/12/2024] [Indexed: 06/22/2024] Open
Abstract
During pulmonary mucormycosis, inhaled sporangiospores adhere to, germinate, and invade airway epithelial cells to establish infection. We provide evidence that HIF1α plays dual roles in airway epithelial cells during Mucorales infection. We observed an increase in HIF1α protein accumulation and increased expression of many known HIF1α-responsive genes during in vitro infection, indicating that HIF1α signaling is activated by Mucorales infection. Inhibition of HIF1α signaling led to a substantial decrease in the ability of R. delemar to invade cultured airway epithelial cells. Transcriptome analysis revealed that R. delemar infection induces the expression of many pro-inflammatory genes whose expression was significantly reduced by HIF1α inhibition. Importantly, pharmacological inhibition of HIF1α increased survival in a mouse model of pulmonary mucormycosis without reducing fungal burden. These results suggest that HIF1α plays two opposing roles during mucormycosis: one that facilitates the ability of Mucorales to invade the host cells and one that facilitates the ability of the host to mount an innate immune response.
Collapse
Affiliation(s)
- Povilas Kavaliauskas
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Yiyou Gu
- Division of Infectious Diseases, The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Naushaba Hasin
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Millipore Sigma, 9900 Blackwell Road, Rockville, MD, 20850, USA
| | - Karen T Graf
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Abdullah Alqarihi
- Division of Infectious Diseases, The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Amol C Shetty
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Carrie McCracken
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Thomas J Walsh
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Center for Innovative Therapeutics and Diagnostics, 6641 West Broad St., Room 100, Richmond, VA, 23220, USA
| | - Ashraf S Ibrahim
- Division of Infectious Diseases, The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
- David Geffen School of Medicine at UCLA, Los Angeles, CA, 90502, USA
| | - Vincent M Bruno
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
8
|
Panda S, Sahu MC, Turuk J, Pati S. Mucormycosis: A Rare disease to Notifiable Disease. Braz J Microbiol 2024; 55:1065-1081. [PMID: 38561499 PMCID: PMC11153412 DOI: 10.1007/s42770-024-01315-z] [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: 05/10/2023] [Accepted: 03/21/2024] [Indexed: 04/04/2024] Open
Abstract
Mucormycosis is the third most frequent invasive mycosis, following candidiasis and aspergillosis. It is frequently neglected due to its rare occurrence; but recently attend the status of notifiable disease due to its higher incidence in both developed and developing nations. India has received global notice since its estimated instances were greater than the global estimated figures. Mucormycosis has several clinical manifestations, including rhino-orbital-cerebral (ROCM), pulmonary, gastrointestinal, cutaneous, renal, and diffuse Mucormycosis. ROCM is the most frequent clinical manifestation in India, although pulmonary mucormycosis is prevalent worldwide. This review also discusses host defenses, pre disposing risk factors and fungal virulence factors that impair host's ability to prevent fungus invasion and disease establishment. The diagnosis of the disease depends on clinical interventions, histological or microbiological procedures along with molecular methods to obtain timely results. But there are still unmet challenges for rapid diagnosis of the disease. Treatment of the disease is achieved by multimodal approaches such as reversal of underlying predisposing factors, rapid administration of antifungals in optimal doses and surgical procedures to remove infected tissues. Liposomal Amphotericin B, Posaconazole and Isavuconazoles are preferred as the first line of treatment procedures. clinical trials. Different studies have improved the existing drug and under clinical trials while several studies predicted the new potential targets as CotH and Ftr1 as shown in infection and in vitro models. Therefore, current scenario demands a multidisciplinary approach is needed to investigate the prevalence, pathogenesis which is highly important for the advancement of rapid diagnosis and effective treatment.
Collapse
Affiliation(s)
- Sunita Panda
- Regional Medical Research Center, Bhubaneswar, Odisha, 751023, India
| | | | - Jyotirmayee Turuk
- Regional Medical Research Center, Bhubaneswar, Odisha, 751023, India.
| | - Sanghamitra Pati
- Regional Medical Research Center, Bhubaneswar, Odisha, 751023, India.
| |
Collapse
|
9
|
Tanwar M, Singh A, Singh TP, Sharma S, Sharma P. Comprehensive Review on the Virulence Factors and Therapeutic Strategies with the Aid of Artificial Intelligence against Mucormycosis. ACS Infect Dis 2024; 10:1431-1457. [PMID: 38682683 DOI: 10.1021/acsinfecdis.4c00082] [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: 05/01/2024]
Abstract
Mucormycosis, a rare but deadly fungal infection, was an epidemic during the COVID-19 pandemic. The rise in cases (COVID-19-associated mucormycosis, CAM) is attributed to excessive steroid and antibiotic use, poor hospital hygiene, and crowded settings. Major contributing factors include diabetes and weakened immune systems. The main manifesting forms of CAM─cutaneous, pulmonary, and the deadliest, rhinocerebral─and disseminated infections elevated mortality rates to 85%. Recent focus lies on small-molecule inhibitors due to their advantages over standard treatments like surgery and liposomal amphotericin B (which carry several long-term adverse effects), offering potential central nervous system penetration, diverse targets, and simpler dosing owing to their small size, rendering the ability to traverse the blood-brain barrier via passive diffusion facilitated by the phospholipid membrane. Adaptation and versatility in mucormycosis are facilitated by a multitude of virulence factors, enabling the pathogen to dynamically respond to various environmental stressors. A comprehensive understanding of these virulence mechanisms is imperative for devising effective therapeutic interventions against this highly opportunistic pathogen that thrives in immunocompromised individuals through its angio-invasive nature. Hence, this Review delineates the principal virulence factors of mucormycosis, the mechanisms it employs to persist in challenging host environments, and the current progress in developing small-molecule inhibitors against them.
Collapse
Affiliation(s)
- Mansi Tanwar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Anamika Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Tej Pal Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Sujata Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Pradeep Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi-110029, India
| |
Collapse
|
10
|
Lax C, Nicolás FE, Navarro E, Garre V. Molecular mechanisms that govern infection and antifungal resistance in Mucorales. Microbiol Mol Biol Rev 2024; 88:e0018822. [PMID: 38445820 PMCID: PMC10966947 DOI: 10.1128/mmbr.00188-22] [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] [Indexed: 03/07/2024] Open
Abstract
SUMMARYThe World Health Organization has established a fungal priority pathogens list that includes species critical or highly important to human health. Among them is the order Mucorales, a fungal group comprising at least 39 species responsible for the life-threatening infection known as mucormycosis. Despite the continuous rise in cases and the poor prognosis due to innate resistance to most antifungal drugs used in the clinic, Mucorales has received limited attention, partly because of the difficulties in performing genetic manipulations. The COVID-19 pandemic has further escalated cases, with some patients experiencing the COVID-19-associated mucormycosis, highlighting the urgent need to increase knowledge about these fungi. This review addresses significant challenges in treating the disease, including delayed and poor diagnosis, the lack of accurate global incidence estimation, and the limited treatment options. Furthermore, it focuses on the most recent discoveries regarding the mechanisms and genes involved in the development of the disease, antifungal resistance, and the host defense response. Substantial advancements have been made in identifying key fungal genes responsible for invasion and tissue damage, host receptors exploited by the fungus to invade tissues, and mechanisms of antifungal resistance. This knowledge is expected to pave the way for the development of new antifungals to combat mucormycosis. In addition, we anticipate significant progress in characterizing Mucorales biology, particularly the mechanisms involved in pathogenesis and antifungal resistance, with the possibilities offered by CRISPR-Cas9 technology for genetic manipulation of the previously intractable Mucorales species.
Collapse
Affiliation(s)
- Carlos Lax
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, Murcia, Spain
| | - Francisco E. Nicolás
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, Murcia, Spain
| | - Eusebio Navarro
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, Murcia, Spain
| | - Victoriano Garre
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, Murcia, Spain
| |
Collapse
|
11
|
Pintye A, Bacsó R, Kovács GM. Trans-kingdom fungal pathogens infecting both plants and humans, and the problem of azole fungicide resistance. Front Microbiol 2024; 15:1354757. [PMID: 38410389 PMCID: PMC10896089 DOI: 10.3389/fmicb.2024.1354757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/23/2024] [Indexed: 02/28/2024] Open
Abstract
Azole antifungals are abundantly used in the environment and play an important role in managing fungal diseases in clinics. Due to the widespread use, azole resistance is an emerging global problem for all applications in several fungal species, including trans-kingdom pathogens, capable of infecting plants and humans. Azoles used in agriculture and clinics share the mode of action and facilitating cross-resistance development. The extensive use of azoles in the environment, e.g., for plant protection and wood preservation, contributes to the spread of resistant populations and challenges using these antifungals in medical treatments. The target of azoles is the cytochrome p450 lanosterol 14-α demethylase encoded by the CYP51 (called also as ERG11 in the case of yeasts) gene. Resistance mechanisms involve mainly the mutations in the coding region in the CYP51 gene, resulting in the inadequate binding of azoles to the encoded Cyp51 protein, or mutations in the promoter region causing overexpression of the protein. The World Health Organization (WHO) has issued the first fungal priority pathogens list (FPPL) to raise awareness of the risk of fungal infections and the increasingly rapid spread of antifungal resistance. Here, we review the main issues about the azole antifungal resistance of trans-kingdom pathogenic fungi with the ability to cause serious human infections and included in the WHO FPPL. Methods for the identification of these species and detection of resistance are summarized, highlighting the importance of these issues to apply the proper treatment.
Collapse
Affiliation(s)
- Alexandra Pintye
- Centre for Agricultural Research, Plant Protection Institute, HUN-REN, Budapest, Hungary
- Department of Plant Anatomy, Institute of Biology, Eötvös Loránd University, Budapest, Hungary
| | - Renáta Bacsó
- Centre for Agricultural Research, Plant Protection Institute, HUN-REN, Budapest, Hungary
| | - Gábor M. Kovács
- Centre for Agricultural Research, Plant Protection Institute, HUN-REN, Budapest, Hungary
- Department of Plant Anatomy, Institute of Biology, Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
12
|
Gullì SP, Hallur V, Kale P, Menezes GA, Russo A, Singla N. From Spores to Solutions: A Comprehensive Narrative Review on Mucormycosis. Diagnostics (Basel) 2024; 14:314. [PMID: 38337830 PMCID: PMC10855476 DOI: 10.3390/diagnostics14030314] [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: 12/29/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Mucormycosis is an infrequent but fatal illness that mainly affects patients with uncontrolled diabetes mellitus, diabetic ketoacidosis, solid and hematologic neoplasms, organ transplantation, chronic steroid intake, prolonged neutropenia, iron overload states, neonatal prematurity, severe malnutrition, and HIV. Many cases were reported across the world recently following the COVID-19 pandemic. Recent research has led to a better understanding of the pathogenesis of the disease, and global guidelines are now available for managing this serious infection. Herein, we comprehensively review the etiological agents, pathogenesis, clinical presentations, diagnosis, and management of mucormycosis.
Collapse
Affiliation(s)
- Sara Palma Gullì
- Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.P.G.); (A.R.)
| | - Vinaykumar Hallur
- Department of Microbiology, All India Institute of Medical Sciences, Bhubaneswar 751019, India
| | - Pratibha Kale
- Department of Clinical Microbiology, Institute of Liver and Biliary Sciences, New Delhi 110070, India;
| | - Godfred Antony Menezes
- Department of Medical Microbiology & Immunology, RAK College of Medical Sciences, Ras Al Khaimah P.O. Box 11172, United Arab Emirates;
| | - Alessandro Russo
- Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.P.G.); (A.R.)
| | - Nidhi Singla
- Department of Microbiology, Government Medical College and Hospital, Chandigarh 160030, India;
| |
Collapse
|
13
|
Ben-Ami R. Experimental Models to Study the Pathogenesis and Treatment of Mucormycosis. J Fungi (Basel) 2024; 10:85. [PMID: 38276032 PMCID: PMC10820959 DOI: 10.3390/jof10010085] [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: 11/28/2023] [Revised: 01/18/2024] [Accepted: 01/21/2024] [Indexed: 01/27/2024] Open
Abstract
Mucormycosis presents a formidable challenge to clinicians and researchers. Animal models are an essential part of the effort to decipher the pathogenesis of mucormycosis and to develop novel pharmacotherapeutics against it. Diverse model systems have been established, using a range of animal hosts, immune and metabolic perturbations, and infection routes. An understanding of the characteristics, strengths, and drawbacks of these models is needed to optimize their use for specific research aims.
Collapse
Affiliation(s)
- Ronen Ben-Ami
- Department of Infectious Diseases, Tel Aviv Sourasky Medical Center, Faculty of Medicine, Tel Aviv University, Tel Aviv 64239, Israel
| |
Collapse
|
14
|
Gwinn KD, Leung MCK, Stephens AB, Punja ZK. Fungal and mycotoxin contaminants in cannabis and hemp flowers: implications for consumer health and directions for further research. Front Microbiol 2023; 14:1278189. [PMID: 37928692 PMCID: PMC10620813 DOI: 10.3389/fmicb.2023.1278189] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/04/2023] [Indexed: 11/07/2023] Open
Abstract
Medicinal and recreational uses of Cannabis sativa, commonly known as cannabis or hemp, has increased following its legalization in certain regions of the world. Cannabis and hemp plants interact with a community of microbes (i.e., the phytobiome), which can influence various aspects of the host plant. The fungal composition of the C. sativa phytobiome (i.e., mycobiome) currently consists of over 100 species of fungi, which includes phytopathogens, epiphytes, and endophytes, This mycobiome has often been understudied in research aimed at evaluating the safety of cannabis products for humans. Medical research has historically focused instead on substance use and medicinal uses of the plant. Because several components of the mycobiome are reported to produce toxic secondary metabolites (i.e., mycotoxins) that can potentially affect the health of humans and animals and initiate opportunistic infections in immunocompromised patients, there is a need to determine the potential health risks that these contaminants could pose for consumers. This review discusses the mycobiome of cannabis and hemp flowers with a focus on plant-infecting and toxigenic fungi that are most commonly found and are of potential concern (e.g., Aspergillus, Penicillium, Fusarium, and Mucor spp.). We review current regulations for molds and mycotoxins worldwide and review assessment methods including culture-based assays, liquid chromatography, immuno-based technologies, and emerging technologies for these contaminants. We also discuss approaches to reduce fungal contaminants on cannabis and hemp and identify future research needs for contaminant detection, data dissemination, and management approaches. These approaches are designed to yield safer products for all consumers.
Collapse
Affiliation(s)
- Kimberly D. Gwinn
- Department of Entomology and Plant Pathology, University of Tennessee, Knoxville, TN, United States
| | - Maxwell C. K. Leung
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ, United States
| | - Ariell B. Stephens
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ, United States
| | - Zamir K. Punja
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
15
|
Alqarihi A, Kontoyiannis DP, Ibrahim AS. Mucormycosis in 2023: an update on pathogenesis and management. Front Cell Infect Microbiol 2023; 13:1254919. [PMID: 37808914 PMCID: PMC10552646 DOI: 10.3389/fcimb.2023.1254919] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
Mucormycosis (MCR) is an emerging and frequently lethal fungal infection caused by the Mucorales family, with Rhizopus, Mucor, and Lichtheimia, accounting for > 90% of all cases. MCR is seen in patients with severe immunosuppression such as those with hematologic malignancy or transplantation, Diabetes Mellitus (DM) and diabetic ketoacidosis (DKA) and immunocompetent patients with severe wounds. The recent SARS COV2 epidemy in India has resulted in a tremendous increase in MCR cases, typically seen in the setting of uncontrolled DM and corticosteroid use. In addition to the diversity of affected hosts, MCR has pleiotropic clinical presentations, with rhino-orbital/rhino-cerebral, sino-pulmonary and necrotizing cutaneous forms being the predominant manifestations. Major insights in MCR pathogenesis have brought into focus the host receptors (GRP78) and signaling pathways (EGFR activation cascade) as well as the adhesins used by Mucorales for invasion. Furthermore, studies have expanded on the importance of iron availability and the complex regulation of iron homeostasis, as well as the pivotal role of mycotoxins as key factors for tissue invasion. The molecular toolbox to study Mucorales pathogenesis remains underdeveloped, but promise is brought by RNAi and CRISPR/Cas9 approaches. Important recent advancements have been made in early, culture-independent molecular diagnosis of MCR. However, development of new potent antifungals against Mucorales remains an unmet need. Therapy of MCR is multidisciplinary and requires a high index of suspicion for initiation of early Mucorales-active antifungals. Reversal of underlying immunosuppression, if feasible, rapid DKA correction and in selected patients, surgical debulking are crucial for improved outcomes.
Collapse
Affiliation(s)
- Abdullah Alqarihi
- Division of Infectious Diseases, The Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles (UCLA) Medical Center, Torrance, CA, United States
| | - Dimitrios P Kontoyiannis
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States
| | - Ashraf S Ibrahim
- Division of Infectious Diseases, The Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles (UCLA) Medical Center, Torrance, CA, United States
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| |
Collapse
|
16
|
Corzo-León DE, Uehling JK, Ballou ER. Rhizopus arrhizus. Trends Microbiol 2023; 31:985-987. [PMID: 37062623 DOI: 10.1016/j.tim.2023.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 04/18/2023]
Affiliation(s)
| | - Jessie K Uehling
- Department of Botany and Plant Pathology, Oregon State University, OR, USA
| | | |
Collapse
|
17
|
Scaife K, Vo TD, Dommels Y, Leune E, Albermann K, Pařenicová L. In silico and in vitro safety assessment of a fungal biomass from Rhizomucor pusillus for use as a novel food ingredient. Food Chem Toxicol 2023; 179:113972. [PMID: 37532172 DOI: 10.1016/j.fct.2023.113972] [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: 05/23/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023]
Abstract
To address the growing world population and reduce the impact of environmental changes on the global food supply, ingredients are being produced using microorganisms to yield sustainable and innovative products. Food ingredients manufactured using modern biotechnology must be produced by non-toxigenic and nonpathogenic production organisms that do not harbor antimicrobial resistance (AMR). Several fungal species represent attractive targets as sources of alternative food products. One such product is a fungal biomass obtained from the fermentation of Rhizomucor pusillus strain CBS 143028. The whole genome sequence of this strain was annotated and subjected to sequence homology searches and in silico phenotype prediction tools to identify genetic elements encoding for protein toxins active via oral consumption, virulence factors associated with pathogenicity, and determinants of AMR. The in silico investigation revealed no genetic elements sharing significant sequence homology with putative virulence factors, protein toxins, or AMR determinants, including the absence of mucoricin, an essential toxin in the pathogenesis of mucormycosis. These in silico findings were corroborated in vitro based on the absence of clinically relevant mycotoxin or antibacterial secondary metabolites. Consequently, it is unlikely that R. pusillis strain CBS 143028 would pose a safety concern for use in food for human consumption.
Collapse
Affiliation(s)
- Kevin Scaife
- Intertek Health Sciences Inc., 2233 Argentia Road, Suite 21, Mississauga, ON, L5N 2X7, Canada.
| | - Trung D Vo
- Intertek Health Sciences Inc., 2233 Argentia Road, Suite 21, Mississauga, ON, L5N 2X7, Canada
| | - Yvonne Dommels
- The Protein Brewery B.V., Goeseelsstraat 10, 4817, MV, Breda, the Netherlands
| | - Elisa Leune
- The Protein Brewery B.V., Goeseelsstraat 10, 4817, MV, Breda, the Netherlands
| | - Kaj Albermann
- Labvantage - Biomax GmbH, Robert-Koch-Str. 2, 82152, Planegg, Germany
| | - Lucie Pařenicová
- The Protein Brewery B.V., Goeseelsstraat 10, 4817, MV, Breda, the Netherlands; BioXact, Böttgerwater 44, 2497, ZJ, Den Haag, Netherlands
| |
Collapse
|
18
|
Lionakis MS, Drummond RA, Hohl TM. Immune responses to human fungal pathogens and therapeutic prospects. Nat Rev Immunol 2023; 23:433-452. [PMID: 36600071 PMCID: PMC9812358 DOI: 10.1038/s41577-022-00826-w] [Citation(s) in RCA: 98] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2022] [Indexed: 01/06/2023]
Abstract
Pathogenic fungi have emerged as significant causes of infectious morbidity and death in patients with acquired immunodeficiency conditions such as HIV/AIDS and following receipt of chemotherapy, immunosuppressive agents or targeted biologics for neoplastic or autoimmune diseases, or transplants for end organ failure. Furthermore, in recent years, the spread of multidrug-resistant Candida auris has caused life-threatening outbreaks in health-care facilities worldwide and raised serious concerns for global public health. Rapid progress in the discovery and functional characterization of inborn errors of immunity that predispose to fungal disease and the development of clinically relevant animal models have enhanced our understanding of fungal recognition and effector pathways and adaptive immune responses. In this Review, we synthesize our current understanding of the cellular and molecular determinants of mammalian antifungal immunity, focusing on observations that show promise for informing risk stratification, prognosis, prophylaxis and therapies to combat life-threatening fungal infections in vulnerable patient populations.
Collapse
Affiliation(s)
- Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Rebecca A Drummond
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Tobias M Hohl
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
19
|
Cánovas-Márquez JT, Lax C, Tahiri G, Navarro E, Nicolás FE, Garre V. Advances in understanding infections caused by the basal fungus Mucor. PLoS Pathog 2023; 19:e1011394. [PMID: 37262085 DOI: 10.1371/journal.ppat.1011394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023] Open
Affiliation(s)
- José T Cánovas-Márquez
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, Murcia, Spain
| | - Carlos Lax
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, Murcia, Spain
| | - Ghizlane Tahiri
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, Murcia, Spain
| | - Eusebio Navarro
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, Murcia, Spain
| | - Francisco E Nicolás
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, Murcia, Spain
| | - Victoriano Garre
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, Murcia, Spain
| |
Collapse
|
20
|
Tayabali K, Pothiwalla H, Narayanan S. Epidemiology of COVID-19-Associated Mucormycosis. CURRENT FUNGAL INFECTION REPORTS 2023; 17:1-20. [PMID: 37360859 PMCID: PMC10155162 DOI: 10.1007/s12281-023-00464-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2023] [Indexed: 06/28/2023]
Abstract
Purpose of the Review To describe the epidemiology and risk factors for Coronavirus disease-19 (COVID-19)-associated mucormycosis (CAM) based on current published literature. Recent Findings COVID-19 is associated with an increased risk of secondary infections. Mucormycosis is an uncommon invasive fungal infection that typically affects people with immunocompromising conditions and uncontrolled diabetes. Treatment of mucormycosis is challenging and is associated with high mortality even with standard care. During the second wave of the COVID 19 pandemic, an abnormally high number of CAM cases were seen particularly in India. Several case series have attempted to describe the risk factors for CAM. Summary A common risk profile identified for CAM includes uncontrolled diabetes and treatment with steroids. COVID-19-induced immune dysregulation as well as some unique pandemic specific risk factors may have played a role.
Collapse
Affiliation(s)
- Khadija Tayabali
- Institute of Human Virology, University of Maryland Medical Center, 725 W Lombard St, S211A, Baltimore, MD 21201 USA
| | | | - Shivakumar Narayanan
- Institute of Human Virology, University of Maryland Medical Center, 725 W Lombard St, S211A, Baltimore, MD 21201 USA
| |
Collapse
|
21
|
Abstract
The respiratory tree maintains sterilizing immunity against human fungal pathogens. Humans inhale ubiquitous filamentous molds and geographically restricted dimorphic fungal pathogens that form small airborne conidia. In addition, pathogenic yeasts, exemplified by encapsulated Cryptococcus species, and Pneumocystis pose significant fungal threats to the lung. Classically, fungal pneumonia occurs in immune compromised individuals, specifically in patients with HIV/AIDS, in patients with hematologic malignancies, in organ transplant recipients, and in patients treated with corticosteroids and targeted biologics that impair fungal immune surveillance in the lung. The emergence of fungal co-infections during severe influenza and COVID-19 underscores the impairment of fungus-specific host defense pathways in the lung by respiratory viruses and by medical therapies to treat viral infections. Beyond life-threatening invasive syndromes, fungal antigen exposure can exacerbate allergenic disease in the lung. In this review, we discuss emerging principles of lung-specific antifungal immunity, integrate the contributions and cooperation of lung epithelial, innate immune, and adaptive immune cells to mucosal barrier immunity, and highlight the pathogenesis of fungal-associated allergenic disease. Improved understanding of fungus-specific immunity in the respiratory tree has paved the way to develop improved diagnostic, pre-emptive, therapeutic, and vaccine approaches for fungal diseases of the lung.
Collapse
Affiliation(s)
- Lena J Heung
- Division of Infectious Diseases, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Darin L Wiesner
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Keyi Wang
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Amariliz Rivera
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Tobias M Hohl
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
22
|
Ma Y, Deng W, Zhang K, Song Y, Zhang L, Shao J, Liu X, Wan Z, Wang X, Li R. Dual RNA-Sequencing and Liquid Chromatography-Mass Spectrometry Unveil Specific Insights on the Pathogenicity of Trichophyton mentagrophytes Complex. J Invest Dermatol 2023; 143:470-479.e6. [PMID: 38295003 DOI: 10.1016/j.jid.2022.08.055] [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: 03/28/2022] [Revised: 07/29/2022] [Accepted: 08/09/2022] [Indexed: 11/22/2022]
Abstract
Trichophyton mentagrophytes is increasingly considered to be a public health hazard because it causes the most severe manifestations of dermatophytosis. In this study, we performed a series of studies to determine the pathogenicity of the T. mentagrophytes complex. We show that the T. mentagrophytes complex interacts with keratinocytes through pattern-recognition receptors‒MAPK/noncanonical NF-κB pathways and that the hyphal form of T. mentagrophytes is responsible for the increased inflammatory responses in keratinocytes. Moreover, SN-38 is likely a toxin of T. mentagrophytes that induces apoptosis in keratinocytes both in vivo and in vitro. Our results explain the severe pathogenicity and destructiveness of T. mentagrophytes observed in the clinic and pave the way for designing novel toxin-directed therapies to improve patient outcomes.
Collapse
Affiliation(s)
- Yubo Ma
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China; Research Center for Medical Mycology, Peking University, Beijing, China; Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China; National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Weiwei Deng
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China; Research Center for Medical Mycology, Peking University, Beijing, China; Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China; National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Kai Zhang
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China; Research Center for Medical Mycology, Peking University, Beijing, China; Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China; National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Yinggai Song
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China; Research Center for Medical Mycology, Peking University, Beijing, China; Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China; National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Lu Zhang
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China; Research Center for Medical Mycology, Peking University, Beijing, China; Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China; National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Jin Shao
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China; Research Center for Medical Mycology, Peking University, Beijing, China; Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China; National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Xiao Liu
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China; Research Center for Medical Mycology, Peking University, Beijing, China; Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China; National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Zhe Wan
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China; Research Center for Medical Mycology, Peking University, Beijing, China; Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China; National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Xiaowen Wang
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China; Research Center for Medical Mycology, Peking University, Beijing, China; Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China; National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Ruoyu Li
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China; Research Center for Medical Mycology, Peking University, Beijing, China; Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China; National Clinical Research Center for Skin and Immune Diseases, Beijing, China.
| |
Collapse
|
23
|
Desai N, Pradhan V, Chougule D, Tiwari S, Mandke C, Yadav RM, Athvale A, Kawle J, Pai V, Pawaskar S, Kharkar H, Bhosale S, Parab A, Ansari S, Kumar KH, Mhashal S, Redkar N, Madkaikar M. Perturbations of immune landscape in COVID-19 associated mucormycosis. Mycoses 2023; 66:226-236. [PMID: 36380699 DOI: 10.1111/myc.13546] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/11/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND A rise in secondary fungal infections during the COVID-19 pandemic necessitates a deeper understanding of the associated immunological perturbations. OBJECTIVES To evaluate the clinical and immunological characteristics observed in patients with COVID-19 associated mucormycosis (CAM) infection. PATIENTS/ METHODS Cases of mucormycosis with or post-COVID-19 infection were compared with cases of acute COVID-19 and convalescent COVID-19. Lymphocyte subsets, cytokines and other laboratory markers were compared between the groups. RESULTS The frequency of proposed risk factors for CAM was diabetes mellitus (77%), recent history of steroid use (69%) and hypoxia during COVID-19 infection (52%). Iron metabolism was dysregulated in CAM patients with low TIBC and total iron. Further, CAM was accompanied with lymphopenia with drastic reduction in B cell counts; however, plasmablasts were not altered. Further, CAM patients had low immunoglobulin levels and antibodies specific to mucor peptide did not increase in CAM suggesting dysfunction in B-cell response. There was increase in activated effector cytotoxic CD8 T cells and NK cells in CAM compared with COVID-19 infection and healthy controls. Among T helper cells, Tregs were reduced and Th-1 frequency was increased in CAM compared with COVID-19 infection. A distinct cytokine signature was evident in CAM with increase in IL-1β, IFN-γ, IL-6, IL-22, IL-17A, IL-10, IL-2, IL-8, IL-7, IL-21 and GM-CSF. CONCLUSION This is the first study on immunophenotyping in CAM suggesting the need for long-term monitoring of B-cell function after SARS-CoV-2 in patients with dysregulated glycaemic control and the possible benefit of therapeutic supplementation with intravenous immunoglobulins in CAM.
Collapse
Affiliation(s)
- Nidhi Desai
- Department of Pediatric Immunology and Leukocyte Biology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Vandana Pradhan
- Department of Clinical & Experimental Immunology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Durga Chougule
- Department of Clinical & Experimental Immunology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Smrati Tiwari
- Department of Medicine, G.S. Medical College, King Edward Memorial Hospital, Mumbai, India
| | - Charuta Mandke
- Department of Ophthalmology, HBT Medical College and Dr R N Cooper Hospital, Mumbai, India
| | - Reetika Malik Yadav
- Department of Pediatric Immunology and Leukocyte Biology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Amita Athvale
- Department of Pulmonary Medicine, G.S. Medical College, King Edward Memorial Hospital, Mumbai, India
| | - Juhi Kawle
- Department of Medicine, G.S. Medical College, King Edward Memorial Hospital, Mumbai, India
| | - Vinayak Pai
- Department of Medicine, G.S. Medical College, King Edward Memorial Hospital, Mumbai, India
| | - Swapnal Pawaskar
- Department of Clinical & Experimental Immunology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Harshada Kharkar
- Department of Clinical & Experimental Immunology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Snehal Bhosale
- Department of Pediatric Immunology and Leukocyte Biology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Ankita Parab
- Department of Pediatric Immunology and Leukocyte Biology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Shazia Ansari
- Department of Ophthalmology, HBT Medical College and Dr R N Cooper Hospital, Mumbai, India
| | - Kinnera Harish Kumar
- Department of Otorhinolaryngology, HBT Medical College and Dr R N Cooper Hospital, Mumbai, India
| | - Shashikant Mhashal
- Department of Otolaryngology, HBT Medical College and Dr R N Cooper Hospital, Mumbai, India
| | - Neelam Redkar
- Department of Medicine, HBT Medical College and Dr R N Cooper Hospital, Mumbai, India
| | - Manisha Madkaikar
- Department of Pediatric Immunology and Leukocyte Biology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| |
Collapse
|
24
|
Brown AJP. Fungal resilience and host-pathogen interactions: Future perspectives and opportunities. Parasite Immunol 2023; 45:e12946. [PMID: 35962618 PMCID: PMC10078341 DOI: 10.1111/pim.12946] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 01/31/2023]
Abstract
We are constantly exposed to the threat of fungal infection. The outcome-clearance, commensalism or infection-depends largely on the ability of our innate immune defences to clear infecting fungal cells versus the success of the fungus in mounting compensatory adaptive responses. As each seeks to gain advantage during these skirmishes, the interactions between host and fungal pathogen are complex and dynamic. Nevertheless, simply compromising the physiological robustness of fungal pathogens reduces their ability to evade antifungal immunity, their virulence, and their tolerance against antifungal therapy. In this article I argue that this physiological robustness is based on a 'Resilience Network' which mechanistically links and controls fungal growth, metabolism, stress resistance and drug tolerance. The elasticity of this network probably underlies the phenotypic variability of fungal isolates and the heterogeneity of individual cells within clonal populations. Consequently, I suggest that the definition of the fungal Resilience Network represents an important goal for the future which offers the clear potential to reveal drug targets that compromise drug tolerance and synergise with current antifungal therapies.
Collapse
Affiliation(s)
- Alistair J P Brown
- Medical Research Council Centre for Medical Mycology at the University of Exeter, Exeter, UK
| |
Collapse
|
25
|
Prajapati J, Rao P, Poojara L, Acharya D, Patel SK, Goswami D, Rawal RM. A Comprehensive in vitro and in silico Assessment on Inhibition of CYP51B and Ergosterol Biosynthesis by Eugenol in Rhizopus oryzae. Curr Microbiol 2023; 80:47. [PMID: 36538133 PMCID: PMC9764306 DOI: 10.1007/s00284-022-03108-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 11/03/2022] [Indexed: 12/24/2022]
Abstract
Mucormycosis, also known as Zygomycosis, is a disease caused by invasive fungi, predominantly Rhizopus species belonging to the Order of Mucorales. Seeing from the chemistry perspective, heterocyclic compounds with an "azole" moiety are widely employed as antifungal agent for minimising the effect of mucormycosis as a prescribed treatment. These azoles serve as non-competitive inhibitors of fungal CYP51B by predominantly binding to its heme moiety, rendering its inhibition. However, long-term usage and abuse of azoles as antifungal medicines has resulted in drug resistance among certain fungal pathogens. Hence, there is an unmet need to find alternative therapeutic compounds. In present study, we used various in vitro tests to investigate the antifungal activity of eugenol against R. oryzae/R. arrhizus, including ergosterol quantification to test inhibition of ergosterol production mediated antifungal action. The minimum inhibitory concentration (MIC) value obtained for eugenol was 512 μg/ml with reduced ergosterol concentration of 77.11 ± 3.25% at MIC/2 concentration. Further, the molecular interactions of eugenol with fungal CYP51B were meticulously studied making use of proteomics in silico study including molecular docking and molecular dynamics simulations that showed eugenol to be strongly interacting with heme in an identical fashion to that shown by azole drugs (in this case, clotrimazole was evaluated). This is the first of a kind study showing the simulation study of eugenol with CYP51B of fungi. This inhibition results in ergosterol synthesis and is also studied and compared with keeping clotrimazole as a reference.
Collapse
Affiliation(s)
- Jignesh Prajapati
- grid.411877.c0000 0001 2152 424XDepartment of Biochemistry and Forensic Science, University School of Sciences, Gujarat University, Ahmedabad, Gujarat 380009 India
| | - Priyashi Rao
- grid.411877.c0000 0001 2152 424XDepartment of Biochemistry and Forensic Science, University School of Sciences, Gujarat University, Ahmedabad, Gujarat 380009 India
| | - Lipi Poojara
- grid.411877.c0000 0001 2152 424XDepartment of Biochemistry and Forensic Science, University School of Sciences, Gujarat University, Ahmedabad, Gujarat 380009 India
| | - Dhaval Acharya
- Department of Microbiology, B N Patel Institute of Paramedical and Sciences, Anand, Gujarat 388001 India
| | - Saumya K. Patel
- grid.411877.c0000 0001 2152 424XDepartment of Botany, Bioinformatics and Climate Change Impacts Management, School of Sciences, Gujarat University, Ahmedabad, Gujarat India
| | - Dweipayan Goswami
- grid.411877.c0000 0001 2152 424XDepartment of Microbiology and Biotechnology, University School of Sciences, Gujarat University, Ahmedabad, Gujarat 380009 India
| | - Rakesh M. Rawal
- grid.411877.c0000 0001 2152 424XDepartment of Biochemistry and Forensic Science, University School of Sciences, Gujarat University, Ahmedabad, Gujarat 380009 India ,grid.411877.c0000 0001 2152 424XDepartment of Life Science, University School of Sciences, Gujarat University, Ahmedabad, Gujarat 380009 India
| |
Collapse
|
26
|
Acosta-España JD, Voigt K. An old confusion: Entomophthoromycosis versus mucormycosis and their main differences. Front Microbiol 2022; 13:1035100. [PMID: 36406416 PMCID: PMC9670544 DOI: 10.3389/fmicb.2022.1035100] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022] Open
Abstract
Fungal diseases were underestimated for many years. And the global burden of fungal infections is substantial and has increased in recent years. Invasive fungal infections have been linked to several risk factors in humans which basically depend on the individual homeostasis of the patients. However, many fungi can infect even apparently healthy people. Knowledge of these pathogens is critical in reducing or stopping morbidity and/or mortality statistics due to fungal pathogens. Successful therapeutic strategies rely on rapid diagnosis of the causative fungal agent and the underlying disease. However, the terminology of the diseases was updated to existing phylogenetic classifications and led to confusion in the definition of mucormycosis, conidiobolomycosis, and basidiobolomycosis, which were previously grouped under the now-uncommon term zygomycosis. Therefore, the ecological, taxonomic, clinical, and diagnostic differences are addressed to optimize the understanding and definition of these diseases. The term "coenocytic hyphomycosis" is proposed to summarize all fungal infections caused by Mucorales and species of Basidiobolus and Conidiobolus.
Collapse
Affiliation(s)
- Jaime David Acosta-España
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany,Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany,Postgraduate Program in Infectious Diseases, School of Medicine, Pontificia Universidad Católica del Ecuador, Quito, Ecuador
| | - Kerstin Voigt
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany,Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany,*Correspondence: Kerstin Voigt,
| |
Collapse
|
27
|
Gurunathan S, Lee AR, Kim JH. Antifungal Effect of Nanoparticles against COVID-19 Linked Black Fungus: A Perspective on Biomedical Applications. Int J Mol Sci 2022; 23:12526. [PMID: 36293381 PMCID: PMC9604067 DOI: 10.3390/ijms232012526] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/16/2022] [Accepted: 10/18/2022] [Indexed: 08/21/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a highly transmissible and pathogenic coronavirus that has caused a 'coronavirus disease 2019' (COVID-19) pandemic in multiple waves, which threatens human health and public safety. During this pandemic, some patients with COVID-19 acquired secondary infections, such as mucormycosis, also known as black fungus disease. Mucormycosis is a serious, acute, and deadly fungal infection caused by Mucorales-related fungal species, and it spreads rapidly. Hence, prompt diagnosis and treatment are necessary to avoid high mortality and morbidity rates. Major risk factors for this disease include uncontrolled diabetes mellitus and immunosuppression that can also facilitate increases in mucormycosis infections. The extensive use of steroids to prevent the worsening of COVID-19 can lead to black fungus infection. Generally, antifungal agents dedicated to medical applications must be biocompatible, non-toxic, easily soluble, efficient, and hypoallergenic. They should also provide long-term protection against fungal growth. COVID-19-related black fungus infection causes a severe increase in fatalities. Therefore, there is a strong need for the development of novel and efficient antimicrobial agents. Recently, nanoparticle-containing products available in the market have been used as antimicrobial agents to prevent bacterial growth, but little is known about their efficacy with respect to preventing fungal growth, especially black fungus. The present review focuses on the effect of various types of metal nanoparticles, specifically those containing silver, zinc oxide, gold, copper, titanium, magnetic, iron, and carbon, on the growth of various types of fungi. We particularly focused on how these nanoparticles can impact the growth of black fungus. We also discussed black fungus co-infection in the context of the global COVID-19 outbreak, and management and guidelines to help control COVID-19-associated black fungus infection. Finally, this review aimed to elucidate the relationship between COVID-19 and mucormycosis.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Ah Reum Lee
- CHA Advanced Research Institute, CHA Medical Center, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea
| | - Jin Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
28
|
Montaño DE, Hartung S, Wich M, Ali R, Jungnickel B, von Lilienfeld-Toal M, Voigt K. The TLR-NF-kB axis contributes to the monocytic inflammatory response against a virulent strain of Lichtheimia corymbifera, a causative agent of invasive mucormycosis. Front Immunol 2022; 13:882921. [PMID: 36311802 PMCID: PMC9608459 DOI: 10.3389/fimmu.2022.882921] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 09/20/2022] [Indexed: 11/29/2022] Open
Abstract
Invasive mucormycosis (IM) is a life-threatening infection caused by the fungal order Mucorales, its diagnosis is often delayed, and mortality rates range from 40-80% due to its rapid progression. Individuals suffering from hematological malignancies, diabetes mellitus, organ transplantations, and most recently COVID-19 are particularly susceptible to infection by Mucorales. Given the increase in the occurrence of these diseases, mucormycosis has emerged as one of the most common fungal infections in the last years. However, little is known about the host immune response to Mucorales. Therefore, we characterized the interaction among L. corymbifera—one of the most common causative agents of IM—and human monocytes, which are specialized phagocytes that play an instrumental role in the modulation of the inflammatory response against several pathogenic fungi. This study covered four relevant aspects of the host-pathogen interaction: i) The recognition of L. corymbifera by human monocytes. ii) The intracellular fate of L. corymbifera. iii) The inflammatory response by human monocytes against the most common causative agents of mucormycosis. iv) The main activated Pattern-Recognition Receptors (PRRs) inflammatory signaling cascades in response to L. corymbifera. Here, we demonstrate that L. corymbifera exhibits resistance to intracellular killing over 24 hours, does not germinate, and inflicts minimal damage to the host cell. Nonetheless, viable fungal spores of L. corymbifera induced early production of the pro-inflammatory cytokine IL-1β, and late release of TNF-α and IL-6 by human monocytes. Moreover, we revealed that IL-1β production predominantly depends on Toll-like receptors (TLRs) priming, especially via TLR4, while TNF-α is secreted via C-type lectin receptors (CTLs), and IL-6 is produced by synergistic activation of TLRs and CTLs. All these signaling pathways lead to the activation of NF-kB, a transcription factor that not only regulates the inflammatory response but also the apoptotic fate of monocytes during infection with L. corymbifera. Collectively, our findings provide new insights into the host-pathogen interactions, which may serve for future therapies to enhance the host inflammatory response to L. corymbifera.
Collapse
Affiliation(s)
- Dolly E. Montaño
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (HKI), Jena, Germany
- Jena Microbial Resource Collection, Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany
| | - Susann Hartung
- Infections in Hematology and Oncology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (HKI), Jena, Germany
| | - Melissa Wich
- Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Jena, Germany
| | - Rida Ali
- Jena Microbial Resource Collection, Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany
| | - Berit Jungnickel
- Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Jena, Germany
| | - Marie von Lilienfeld-Toal
- Infections in Hematology and Oncology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (HKI), Jena, Germany
- Department of Hematology and Medical Oncology, Jena University Hospital, Jena, Germany
| | - Kerstin Voigt
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (HKI), Jena, Germany
- Jena Microbial Resource Collection, Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany
- *Correspondence: Kerstin Voigt,
| |
Collapse
|
29
|
Radotra B, Challa S. Pathogenesis and Pathology of COVID-Associated Mucormycosis: What Is New and Why. CURRENT FUNGAL INFECTION REPORTS 2022; 16:206-220. [PMID: 36193101 PMCID: PMC9520103 DOI: 10.1007/s12281-022-00443-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2022] [Indexed: 12/02/2022]
Abstract
Purpose of Review There is global increase in the incidence of mucormycosis. However, a sudden increase in the COVID-associated mucormycosis (CAM) was noted, particularly in India, during the second wave of the COVID-19 pandemic. The interplay of factors involved in the pathogenesis is complex. In this review, the influence of pre-existing disease, exaggerated risk factors, altered milieu due to COVID-19 itself and the consequences of its treatment on the host pathogen interactions leading to the disease and morphology of the fungus will be highlighted. Recent Findings Hyperglycemia, acidosis, available free iron, lowered host defenses, and the fungal virulence factors promote the growth of Mucorales. There is a high background prevalence of diabetes mellitus (DM) in India. Uncontrolled or undiagnosed DM, COVID-19 itself, and inappropriate administration of corticosteroids in high doses and for prolonged periods result in hyperglycemia. Diabetic ketoacidosis (DKA) and metabolic acidosis due to hypoxia or renal failure contribute to acidic pH and dissociate bound iron from serum proteins. The host defenses are lowered due to COVID-19-induced immune dysregulation, hyperglycemia itself, and administration of corticosteroids and immune suppressants for the treatment of COVID-19. The altered metabolic milieu in the local microenvironment of nose and paranasal sinuses (PNS) promotes specific interaction of glucose-regulated protein-78 (GRP-78) on host cells with spore coat protein homologue (CotH 3) on Mucorales resulting in rhino-orbito-cerebral mucormycosis (ROCM) as the predominant clinical form in CAM. The pathology is extensive soft tissue involvement with angioinvasion and perineural invasion. Melanized hyphae and sporangia were seen on histopathology, which is unique to CAM. While many factors favor the growth of Mucorales in CAM, hyperglycemia, hyperferritinemia, and administration of hyperbaric oxygen result in reactive oxygen species (ROS) and inadequate humidification results in dehydration. Melanization is possibly the adaptive and protective mechanism of Mucorales to escape the unfavorable conditions due to the treatment of COVID-19. Summary High background prevalence of DM, inappropriate administration of corticosteroids and immune dysregulation due to COVID-19 favor the growth of Mucorales in CAM. Melanization of Mucorales hyphae and sporangia on histopathology probably represent adaptive and protective mechanism due to the treatment with hyperbaric oxygen with inadequate humidification as well as the metabolic alterations.
Collapse
Affiliation(s)
- Bishan Radotra
- Department of Histopathology, Group “C” Departments, Postgraduate Institute of Medical Education & Research, Chandigarh, 160012 India
| | - Sundaram Challa
- Department of Pathology and Lab Medicine, Basavatarakam Indo-American Cancer Hospital & Research Institute, Hyderabad, Telangana State 50034 India
| |
Collapse
|
30
|
Lipase-Catalyzed Synthesis, Antioxidant Activity, Antimicrobial Properties and Molecular Docking Studies of Butyl Dihydrocaffeate. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27155024. [PMID: 35956977 PMCID: PMC9370587 DOI: 10.3390/molecules27155024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 11/22/2022]
Abstract
Green chemistry approaches, such as lipase-catalyzed esterification, are promising methods for obtaining valuable chemical compounds. In the case of the use of lipases, unlike in aqueous environments, the processes of the ester bond formations are encountered in organic solvents. The aim of the current research was to carry out the lipase-catalyzed synthesis of an ester of dihydrocaffeic acid. The synthesized compound was then evaluated for antioxidant and antimicrobial activities. However, the vast majority of its antioxidant activity was retained, which was demonstrated by means of DPPH· (2,2-diphenyl-1-picrylhydrazyl) and CUPRAC (cupric ion reducing antioxidant capacity) methods. Regarding its antimicrobial properties, the antifungal activity against Rhizopus oryzae is worth mentioning. The minimum inhibitory and fungicidal concentrations were 1 and 2 mM, respectively. The high antifungal activity prompted the use of molecular docking studies to verify potential protein targets for butyl ester of dihydrocaffeic ester. In the case of one fungal protein, namely 14-α sterol demethylase B, it was observed that the ester had comparable binding energy to the triazole medication, isavuconazole, but the interacted amino acid residues were different.
Collapse
|
31
|
Abstract
Coronavirus disease 2019 (COVID-19)-associated invasive fungal infections are an important complication in a substantial number of critically ill, hospitalized patients with COVID-19. Three groups of fungal pathogens cause co-infections in COVID-19: Aspergillus, Mucorales and Candida species, including Candida auris. Here we review the incidence of COVID-19-associated invasive fungal infections caused by these fungi in low-, middle- and high-income countries. By evaluating the epidemiology, clinical risk factors, predisposing features of the host environment and immunological mechanisms that underlie the pathogenesis of these co-infections, we set the scene for future research and development of clinical guidance. Hoenigl and colleagues review the epidemiology, immunology and clinical risk factors contributing to COVID-19-associated fungal infections.
Collapse
|
32
|
Hoenigl M, Seidel D, Carvalho A, Rudramurthy SM, Arastehfar A, Gangneux JP, Nasir N, Bonifaz A, Araiza J, Klimko N, Serris A, Lagrou K, Meis JF, Cornely OA, Perfect JR, White PL, Chakrabarti A. The emergence of COVID-19 associated mucormycosis: a review of cases from 18 countries. THE LANCET. MICROBE 2022; 3:e543-e552. [PMID: 35098179 PMCID: PMC8789240 DOI: 10.1016/s2666-5247(21)00237-8] [Citation(s) in RCA: 235] [Impact Index Per Article: 78.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Reports of COVID-19-associated mucormycosis have been increasing in frequency since early 2021, particularly among patients with uncontrolled diabetes. Patients with diabetes and hyperglycaemia often have an inflammatory state that could be potentiated by the activation of antiviral immunity to SARS-CoV2, which might favour secondary infections. In this Review, we analysed 80 published and unpublished cases of COVID-19-associated mucormycosis. Uncontrolled diabetes, as well as systemic corticosteroid treatment, were present in most patients with COVID-19-associated mucormycosis, and rhino-orbital cerebral mucormycosis was the most frequent disease. Mortality was high at 49%, which was particularly due to patients with pulmonary or disseminated mucormycosis or cerebral involvement. Furthermore, a substantial proportion of patients who survived had life-changing morbidities (eg, loss of vision in 46% of survivors). Our Review indicates that COVID-19-associated mucormycosis is associated with high morbidity and mortality. Diagnosis of pulmonary mucormycosis is particularly challenging, and might be frequently missed in India.
Collapse
Affiliation(s)
- Martin Hoenigl
- Division of Infectious Diseases, ECMM Center of Excellence for Medical Mycology, Medical University of Graz, Graz, Austria
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Clinical and Translational Fungal Working Group, University of California San Diego, La Jolla, CA, USA
| | - Danila Seidel
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
- Department of Internal Medicine, ECMM Center of Excellence for Medical Mycology, University of Cologne, Cologne, Germany
- German Centre for Infection Research, Partner Site Bonn-Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- PT Government Associate Laboratory, Guimarães, Portugal
| | - Shivaprakash M Rudramurthy
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Amir Arastehfar
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Jean-Pierre Gangneux
- Environnement et Travail, Univ Rennes, CHU Rennes, Inserm, Institut de Recherche en Santé, Rennes, France
| | - Nosheen Nasir
- Section of Adult Infectious Diseases, Department of Medicine, Aga Khan University Karachi, Karachi, Pakistan
| | - Alexandro Bonifaz
- Dermatology Service, Hospital General De México Dr Eduardo Liceaga, Mexico City, Mexico
| | - Javier Araiza
- Dermatology Service, Hospital General De México Dr Eduardo Liceaga, Mexico City, Mexico
| | - Nikolai Klimko
- Department of Clinical Mycology, Allergy and Immunology, North Western State Medical University named after II Mechnikov, St Petersburg, Russia
| | - Alexandra Serris
- Department of Infectious Diseases, Necker-Enfants Malades University Hospital, Paris, France
| | - Katrien Lagrou
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Department of Laboratory Medicine and National Reference Centre for Mycosis, ECMM Center of Excellence for Medical Mycology, University Hospitals Leuven, Leuven, Belgium
| | - Jacques F Meis
- Department of Medical Microbiology and Infectious Diseases, ECMM Center of Excellence for Medical Mycology, Radboud University Medical Center, Canisius Wilhelmina Hospital, Nijmegen, Netherlands
- Center of Expertise in Mycology, Radboud University Medical Center, Canisius Wilhelmina Hospital, Nijmegen, Netherlands
- Bioprocess Engineering and Biotechnology Graduate Program, Federal University of Paraná, Curitiba, Brazil
| | - Oliver A Cornely
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
- Department of Internal Medicine, ECMM Center of Excellence for Medical Mycology, University of Cologne, Cologne, Germany
- German Centre for Infection Research, Partner Site Bonn-Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Clinical Trials Centre Cologne, ZKS Köln, University of Cologne, Cologne, Germany
| | - John R Perfect
- Division of Infectious Diseases, Duke University Medical Center, Durham, NC, USA
| | - P Lewis White
- Public Health Wales Mycology Reference Laboratory, UHW, Cardiff, UK
| | - Arunaloke Chakrabarti
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
33
|
Current Treatment Options for COVID-19 Associated Mucormycosis: Present Status and Future Perspectives. J Clin Med 2022; 11:jcm11133620. [PMID: 35806905 PMCID: PMC9267579 DOI: 10.3390/jcm11133620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/12/2022] [Accepted: 05/17/2022] [Indexed: 12/04/2022] Open
Abstract
Mucormycosis has become increasingly associated with COVID-19, leading to the use of the term “COVID-19 associated mucormycosis (CAM)”. Treatment of CAM is challenging due to factors such as resistance to many antifungals and underlying co-morbidities. India is particularly at risk for this disease due to the large number of patients with COVID-19 carrying comorbidities that predispose them to the development of mucormycosis. Additionally, mucormycosis treatment is complicated due to the atypical symptoms and delayed presentation after the resolution of COVID-19. Since this disease is associated with increased morbidity and mortality, early identification and diagnosis are desirable to initiate a suitable combination of therapies and control the disease. At present, the first-line treatment involves Amphotericin B and surgical debridement. To overcome limitations associated with surgery (invasive, multiple procedures required) and amphotericin B (toxicity, extended duration and limited clinical success), additional therapies can be utilized as adjuncts or alternatives to reduce treatment duration and improve prognosis. This review discusses the challenges associated with treating CAM and the critical aspects for controlling this invasive fungal infection—early diagnosis and initiation of therapy, reversal of risk factors, and adoption of a multipronged treatment strategy. It also details the various therapeutic options (in vitro, in vivo and human case reports) that have been used for the treatment of CAM.
Collapse
|
34
|
Secretion of the siderophore rhizoferrin is regulated by the cAMP-PKA pathway and is involved in the virulence of Mucor lusitanicus. Sci Rep 2022; 12:10649. [PMID: 35739200 PMCID: PMC9226013 DOI: 10.1038/s41598-022-14515-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/08/2022] [Indexed: 11/08/2022] Open
Abstract
Mucormycosis is a fungal infection caused by Mucorales, with a high mortality rate. However, only a few virulence factors have been described in these organisms. This study showed that deletion of rfs, which encodes the enzyme for the biosynthesis of rhizoferrin, a siderophore, in Mucor lusitanicus, led to a lower virulence in diabetic mice and nematodes. Upregulation of rfs correlated with the increased toxicity of the cell-free supernatants of the culture broth (SS) obtained under growing conditions that favor oxidative metabolism, such as low glucose levels or the presence of H2O2 in the culture, suggesting that oxidative metabolism enhances virulence through rhizoferrin production. Meanwhile, growing M. lusitanicus in the presence of potassium cyanide, N-acetylcysteine, a higher concentration of glucose, or exogenous cAMP, or the deletion of the gene encoding the regulatory subunit of PKA (pkaR1), correlated with a decrease in the toxicity of SS, downregulation of rfs, and reduction in rhizoferrin production. These observations indicate the involvement of the cAMP-PKA pathway in the regulation of rhizoferrin production and virulence in M. lusitanicus. Moreover, rfs upregulation was observed upon macrophage interaction or during infection with spores in mice, suggesting a pivotal role of rfs in M. lusitanicus infection.
Collapse
|
35
|
Ribotoxic Proteins, Known as Inhibitors of Protein Synthesis, from Mushrooms and Other Fungi According to Endo's Fragment Detection. Toxins (Basel) 2022; 14:toxins14060403. [PMID: 35737065 PMCID: PMC9227437 DOI: 10.3390/toxins14060403] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/06/2022] [Accepted: 06/10/2022] [Indexed: 12/15/2022] Open
Abstract
rRNA N-glycosylases (EC 3.2.2.22) remove a specific adenine (A4324, rat 28S rRNA) in the sarcin ricin loop (SRL) involved into ribosome interaction with elongation factors, causing the inhibition of translation, for which they are known as plant 'ribosome inactivating proteins' (RIPs). However, protein synthesis inactivation could be the result of other enzymes, which often have rRNA as the target. In this scenario, Endo's assay is the most used method to detect the enzymes that are able to hydrolyze a phosphodiester bond or cleave a single N-glycosidic bond (rRNA N-glycosylases). Indeed, the detection of a diagnostic fragment from rRNA after enzymatic action, with or without acid aniline, allows one to discriminate between the N-glycosylases or hydrolases, which release the β-fragment after acid aniline treatment or α-fragment without acid aniline treatment, respectively. This assay is of great importance in the mushroom kingdom, considering the presence of enzymes that are able to hydrolyze phosphodiester bonds (e.g., ribonucleases, ribotoxins and ribotoxin-like proteins) or to remove a specific adenine (rRNA N-glycosylases). Thus, here we used the β-fragment experimentally detected by Endo's assay as a hallmark to revise the literature available on enzymes from mushrooms and other fungi, whose action consists of protein biosynthesis inhibition.
Collapse
|
36
|
Clinical Manifestations of Pulmonary Mucormycosis in Recipients of Allogeneic Hematopoietic Stem Cell Transplantation: A 21-Case Series Report and Literature Review. Can Respir J 2022; 2022:1237125. [PMID: 35692949 PMCID: PMC9184213 DOI: 10.1155/2022/1237125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 05/09/2022] [Accepted: 05/13/2022] [Indexed: 11/17/2022] Open
Abstract
Introduction Mucormycosis is a rare, invasive disease caused by opportunistic pathogens related to the Mucorales order with high fatality rates in immunocompromised hosts, especially in recipients of allogeneic hematopoietic stem cell transplantation (allo-HSCT). Diagnosis and treatment of pulmonary mucormycosis in recipients of allo-HSCT remains challenging. Purpose The aim of this study is to summarize and analyze the clinical features of pulmonary mucormycosis in recipients of allo-HSCT to explore further clinical research directions for this rare fungal infection in the particular populations. Methods We retrospectively reviewed pulmonary mucormycosis in patients who received allo-HSCT in our hospital from January 2010 to December 2020. A total of 21 patients fulfilled the diagnostic criteria for pulmonary mucormycosis according to the European Organization for Research and Treatment of Cancer and Mycoses Study Group (EORTC/MSG) criteria. Demographic and clinical data, mycological and histopathological records, and treatment and prognosis data were collected. Clinical variables were compared between survivors and nonsurvivors. The survival days of patients with and without graft-versus-host disease (GVHD) and hemoptysis were compared separately. Results Most of the recipients of allo-HSCT were male patients with a mean age of 43 years. Acute myeloid leukemia (AML) was the most common primary hematologic malignancy. Extrapulmonary involvement accounted for 28.6%, of the cases, including central nervous system (n = 5) and skin and soft tissue (n = 1). The median time to infection was 96 days after allo-HSCT. Clinical presentations were nonspecific, including fever (76.2%) and cough (85.7%), as well as dyspnea (19.0%), chest pain (38.1%), and hemoptysis (61.9%). Ground-glass infiltrates (95.0%) and nodules/masses (80%) were the most common radiographic patterns on chest CT. The most common pathogen was Rhizopus (63.2%), and breakthrough infection accounted for 90.5%. Fifteen of the patients died within one year, and the median time from diagnosis to death was 47 days. Conclusion Mucormycosis is a fatal infection disease. Opportunistic infections in recipients of allo-HSCT are mainly breakthrough infections and may have a seasonal distribution (summer and autumn) and more cases of death in autumn. The marked reversed halo sign can be seen both in the initial stage of infection and after antifungal treatment. In our case series, patients with pulmonary mucormycosis with extrapulmonary involvement 100% died within one year. There are more patients with GVHD before infection and hemoptysis in nonsurvivors than survivors within 100 days. Patients with GVHD before infection and hemoptysis have a shorter survival time than those without.
Collapse
|
37
|
Abstract
INTRODUCTION Disease due to pulmonary infection with Aspergillus, and other emerging opportunistic fungi remains a significant unmet need. Existing antifungal medicines are predominantly dosed either orally or systemically, but because of limited exposure to the lung lumen, adverse events, and problematic drug-drug interactions, inhaled treatment could provide an attractive option. AREA COVERED This review summarizes 1) the limitations of current antifungal therapy, 2) the beneficial effects of inhaled antifungal agents, 3) the clinical development of inhaled antifungal triazoles (repurposed with an innovative inhalation system or a novel inhaled agent) for the treatment of pulmonary fungal infections, and 4) the difficulties and challenges of inhaled antifungal agent development. Regrettably, details of novel inhaled devices or formulations were not covered. EXPERT OPINION Inhaled antifungal treatment could provide an attractive option by shifting the risk benefit ratio of treatment favorably. Preclinical and clinical studies with inhaled antifungal agents (off-label use) are encouraging so far. New inhaled antifungal triazoles are well tolerated in early clinical studies and warrant further clinical development. However, challenges remain and many unaddressed issues including required preclinical studies, appropriate clinical design, pharmacokinetics, delivery system(s) and regulatory process need to be resolved. Early communication with regulatory authorities is therefore recommended.
Collapse
Affiliation(s)
- Kazuhiro Ito
- Respiratory Molecular Medicine, Genomic and Environmental Medicine section, National Heart and Lung Institute, Imperial College, London, UK
| |
Collapse
|
38
|
Dong N, Jordan AE, Shen X, Wu X, Guo X, Zhao H, Wang Y, Wang D, Fang Q. Rhino-Orbital Cerebral Mucormycosis in a Patient With Diabetic Ketoacidosis: A Case Report and Literature Review. Front Neurol 2022; 13:815902. [PMID: 35599740 PMCID: PMC9114505 DOI: 10.3389/fneur.2022.815902] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/21/2022] [Indexed: 12/14/2022] Open
Abstract
Background Rhino-orbital cerebral mucormycosis (ROCM) is a rare, invasive, and fatal fungal disease. Due to the lack of specific clinical manifestations and adequate auxiliary examinations, patients are easily misdiagnosed in the early stage. Early diagnosis and timely therapy are essential for successful treatment. Case Report We report a 68-year-old man with diabetic ketoacidosis, presented with orbital apex syndrome (OAS), fever, and pansinusitis, which progressively worsened to death only 4 days after admission. It was finally confirmed as a fungal Rhizopus arrhizus infection by metagenomics cell-free DNA next-generation sequencing (mNGS) testing. Conclusion Orbital apex syndrome could be the initial presentation for mucormycosis. Thus, it is necessary to evaluate the presence of mucormycosis in patients with OAS, especially in diabetic or immunosuppressed hosts, and mNGS testing and timely antifungal therapy should be strongly recommended in highly suspected cases.
Collapse
Affiliation(s)
- Nan Dong
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Neurology, Suzhou Industrial Park Xinghai Hospital, Suzhou, China
| | - Ashly E. Jordan
- Independent Research Epidemiologist, New York, NY, United States
| | - Xiaozhu Shen
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xuan Wu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xianghong Guo
- Department of Neurology, Suzhou Industrial Park Xinghai Hospital, Suzhou, China
| | - Hongru Zhao
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yajuan Wang
- Genoxor Medical Science and Technology Inc., Shanghai, China
| | - Dapeng Wang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Fang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
39
|
Taking a Closer Look: Clinical and Histopathological Characteristics of Culture-Positive versus Culture-Negative Pulmonary Mucormycosis. J Fungi (Basel) 2022; 8:jof8040380. [PMID: 35448611 PMCID: PMC9030790 DOI: 10.3390/jof8040380] [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: 03/09/2022] [Revised: 04/01/2022] [Accepted: 04/03/2022] [Indexed: 02/04/2023] Open
Abstract
The cultural recovery of Mucorales from hyphae-laden tissue is poor, and the clinical implications of culture positivity are scarcely studied. Therefore, we compared clinical and histopathological characteristics of culture-positive and culture-negative histology-proven pulmonary mucormycosis cases among cancer patients. Histology specimens were blindly reviewed by a thoracic pathologist and graded on four histopathologic features: hyphal quantity, tissue necrosis, tissue invasion, and vascular invasion. Twenty cases with a corresponding fungal culture were identified; five were culture-positive, and fifteen were culture-negative. Although no statistically significant differences were found, culture-positive patients were more likely to exhibit a high burden of necrosis and have a high burden of hyphae but tended to have less vascular invasion than culture-negative patients. In terms of clinical characteristics, culture-positive patients were more likely to have acute myeloid leukemia (60% vs. 27%, p = 0.19), a history of hematopoietic cell transplant (80% vs. 53%, p = 0.31), severe lymphopenia (absolute lymphocyte count ≤ 500/µL, 100% vs. 73%, p = 0.36), and monocytopenia (absolute monocyte count ≤100/µL, 60% vs. 20%, p = 0.11). Forty-two-day all-cause mortality was comparable between culture-positive and culture-negative patients (60% and 53%, p = 0.80). This pilot study represents the first comprehensive histopathological scoring method to examine the relationship between histopathologic features, culture positivity, and clinical features of pulmonary mucormycosis.
Collapse
|
40
|
Garre V. Recent Advances and Future Directions in the Understanding of Mucormycosis. Front Cell Infect Microbiol 2022; 12:850581. [PMID: 35281441 PMCID: PMC8907824 DOI: 10.3389/fcimb.2022.850581] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/04/2022] [Indexed: 11/13/2022] Open
Abstract
Mucormycosis is an emerging infection caused by fungi of the order Mucorales that has recently gained public relevance due to the high incidence among COVID-19 patients in some countries. The reduced knowledge about Mucorales pathogenesis is due, in large part, to the historically low interest for these fungi fostered by their reluctance to be genetically manipulated. The recent introduction of more tractable genetic models together with an increasing number of available whole genome sequences and genomic analyses have improved our understanding of Mucorales biology and mucormycosis in the last ten years. This review summarizes the most significant advances in diagnosis, understanding of the innate and acquired resistance to antifungals, identification of new virulence factors and molecular mechanisms involved in the infection. The increased awareness about the disease and the recent successful genetic manipulation of previous intractable fungal models using CRISPR-Cas9 technology are expected to fuel the characterization of Mucorales pathogenesis, facilitating the development of effective treatments to fight this deadly infection.
Collapse
Affiliation(s)
- Victoriano Garre
- Department of Genetics and Microbiology, Faculty of Biology, University of Murcia, Murcia, Spain
| |
Collapse
|
41
|
Drautz-Moses DI, Luhung I, Gusareva ES, Kee C, Gaultier NE, Premkrishnan BNV, Lee CF, Leong ST, Park C, Yap ZH, Heinle CE, Lau KJX, Purbojati RW, Lim SBY, Lim YH, Kutmutia SK, Aung NW, Oliveira EL, Ng SG, Dacanay J, Ang PN, Spence S, Phung WJ, Wong A, Kennedy RJ, Kalsi N, Sasi SP, Chandrasekaran L, Uchida A, Junqueira ACM, Kim HL, Hankers R, Feuerle T, Corsmeier U, Schuster SC. Vertical stratification of the air microbiome in the lower troposphere. Proc Natl Acad Sci U S A 2022; 119:e2117293119. [PMID: 35131944 PMCID: PMC8851546 DOI: 10.1073/pnas.2117293119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/11/2021] [Indexed: 12/11/2022] Open
Abstract
The troposphere constitutes the final frontier of global ecosystem research due to technical challenges arising from its size, low biomass, and gaseous state. Using a vertical testing array comprising a meteorological tower and a research aircraft, we conducted synchronized measurements of meteorological parameters and airborne biomass (n = 480) in the vertical air column up to 3,500 m. The taxonomic analysis of metagenomic data revealed differing patterns of airborne microbial community composition with respect to time of day and height above ground. The temporal and spatial resolution of our study demonstrated that the diel cycle of airborne microorganisms is a ground-based phenomenon that is entirely absent at heights >1,000 m. In an integrated analysis combining meteorological and biological data, we demonstrate that atmospheric turbulence, identified by potential temperature and high-frequency three-component wind measurements, is the key driver of bioaerosol dynamics in the lower troposphere. Multivariate regression analysis shows that at least 50% of identified airborne microbial taxa (n = ∼10,000) are associated with either ground or height, allowing for an understanding of dispersal patterns of microbial taxa in the vertical air column. Due to the interconnectedness of atmospheric turbulence and temperature, the dynamics of microbial dispersal are likely to be impacted by rising global temperatures, thereby also affecting ecosystems on the planetary surface.
Collapse
Affiliation(s)
- Daniela I Drautz-Moses
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Irvan Luhung
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Elena S Gusareva
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
- The Asian School of the Environment, Nanyang Technological University, Singapore 637459
| | - Carmon Kee
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Nicolas E Gaultier
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | | | - Choou Fook Lee
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - See Ting Leong
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Changsook Park
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Zhei Hwee Yap
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Cassie E Heinle
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Kenny J X Lau
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Rikky W Purbojati
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Serene B Y Lim
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Yee Hui Lim
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Shruti Ketan Kutmutia
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Ngu War Aung
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Elaine L Oliveira
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Soo Guek Ng
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Justine Dacanay
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Poh Nee Ang
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Sam Spence
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Wen Jia Phung
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Anthony Wong
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Ryan J Kennedy
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Namrata Kalsi
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Santhi Puramadathil Sasi
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Lakshmi Chandrasekaran
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Akira Uchida
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Ana Carolina M Junqueira
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, Brazil
| | - Hie Lim Kim
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
- The Asian School of the Environment, Nanyang Technological University, Singapore 637459
| | - Rudolf Hankers
- Institute of Flight Guidance, Technische Universität, 38108 Braunschweig, Germany
| | - Thomas Feuerle
- Institute of Flight Guidance, Technische Universität, 38108 Braunschweig, Germany
| | - Ulrich Corsmeier
- Institute of Meteorology and Climate Research, Karlsruhe Institute of Technology, 76021 Karlsruhe, Germany
| | - Stephan C Schuster
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551;
| |
Collapse
|
42
|
Madanagopal P, Ramprabhu N, Jagadeesan R. In silico prediction and structure-based multitargeted molecular docking analysis of selected bioactive compounds against mucormycosis. BULLETIN OF THE NATIONAL RESEARCH CENTRE 2022; 46:24. [PMID: 35125861 PMCID: PMC8802264 DOI: 10.1186/s42269-022-00704-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/13/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND During the second wave of the COVID-19 pandemic, an unusual increase in cases of mucormycosis was observed in India, owing to immunological dysregulation caused by the SARS-CoV-2 and the use of broad-spectrum antibiotics, particularly in patients with poorly controlled diabetes with ketoacidosis to have contributed to the rise, and it has been declared an epidemic in several states of India. Because of the black colouring of dead and dying tissue caused by the fungus, it was dubbed "black fungus" by several Indian media outlets. In this study, attempts were taken to unmask novel therapeutic options to treat mucormycosis disease. Rhizopus species is the primary fungi responsible for 70% of mucormycosis cases. RESULTS We chose three important proteins from the Rhizopus delemar such as CotH3, Lanosterol 14 alpha-demethylase and Mucoricin which plays a crucial role in the virulence of Mucorales. Initially, we explored the physiochemical, structural and functional insights of proteins and later using AutoDock Vina, we applied computational protein-ligand binding modelling to perform a virtual screening around 300 selected compounds against these three proteins, including FDA-approved drugs, FDA-unapproved drugs, investigational-only drugs and natural bioactive compounds. ADME parameters, toxicity risk and biological activity of those compounds were approximated via in silico methods. Our computational studies identified six ligands as potential inhibitors against Rhizopus delemar, including 12,28-Oxamanzamine A, vialinin B and deoxytopsentin for CotH3; pramiconazole and saperconazole for Lanosterol 14 alpha-demethylase; and Hesperidin for Mucoricin. Interestingly, 12,28-Oxamanzamine A showed a maximum binding affinity with all three proteins (CotH3: - 10.2 kcal/mol Lanosterol 14 alpha-demethylase: - 10.9 kcal/mol Mucoricin: - 8.6 kcal/mol). CONCLUSIONS In summary, our investigation identified 12,28-Oxamanzamine A, vialinin B, deoxytopsentin, pramiconazole, saperconazole and hesperidin as potent bioactive compounds for treating mucormycosis that may be considered for further optimisation techniques and in vitro and in vivo studies. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1186/s42269-022-00704-4.
Collapse
Affiliation(s)
- Premnath Madanagopal
- Department of Biotechnology, Alagappa College of Technology, Anna University, Chennai, India
| | - Nagarjun Ramprabhu
- Department of Biotechnology, Alagappa College of Technology, Anna University, Chennai, India
| | - Rahul Jagadeesan
- Department of Biotechnology, Alagappa College of Technology, Anna University, Chennai, India
| |
Collapse
|
43
|
Kiselev A, San Clemente H, Camborde L, Dumas B, Gaulin E. A Comprehensive Assessment of the Secretome Responsible for Host Adaptation of the Legume Root Pathogen Aphanomyces euteiches. J Fungi (Basel) 2022; 8:88. [PMID: 35050028 PMCID: PMC8780586 DOI: 10.3390/jof8010088] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/09/2022] [Accepted: 01/14/2022] [Indexed: 01/27/2023] Open
Abstract
The soil-borne oomycete pathogen Aphanomyces euteiches causes devastating root rot diseases in legumes such as pea and alfalfa. The different pathotypes of A. euteiches have been shown to exhibit differential quantitative virulence, but the molecular basis of host adaptation has not yet been clarified. Here, we re-sequenced a pea field reference strain of A. euteiches ATCC201684 with PacBio long-reads and took advantage of the technology to generate the mitochondrial genome. We identified that the secretome of A. euteiches is characterized by a large portfolio of secreted proteases and carbohydrate-active enzymes (CAZymes). We performed Illumina sequencing of four strains of A. euteiches with contrasted specificity to pea or alfalfa and found in different geographical areas. Comparative analysis showed that the core secretome is largely represented by CAZymes and proteases. The specific secretome is mainly composed of a large set of small, secreted proteins (SSP) without any predicted functional domain, suggesting that the legume preference of the pathogen is probably associated with unknown functions. This study forms the basis for further investigations into the mechanisms of interaction of A. euteiches with legumes.
Collapse
Affiliation(s)
| | | | | | | | - Elodie Gaulin
- Laboratoire de Recherche en Sciences Végétales (LRSV), Université de Toulouse, CNRS, UPS, Toulouse INP, 31320 Toulouse, France; (A.K.); (H.S.C.); (L.C.); (B.D.)
| |
Collapse
|
44
|
Lax C, Navarro-Mendoza MI, Pérez-Arques C, Navarro E, Nicolás FE, Garre V. Stable and reproducible homologous recombination enables CRISPR-based engineering in the fungus Rhizopus microsporus. CELL REPORTS METHODS 2021; 1:100124. [PMID: 35475217 PMCID: PMC9017206 DOI: 10.1016/j.crmeth.2021.100124] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/20/2021] [Accepted: 11/09/2021] [Indexed: 04/22/2023]
Abstract
Mucormycosis is a lethal and emerging disease that has lacked a genetic model fulfilling both high virulence and the possibility of performing stable and reproducible gene manipulation by homologous recombination (HR). Here, we developed a new methodology to successfully perform HR in Rhizopus microsporus. We isolated an uracil auxotrophic recipient strain and optimized the critical steps in the genetic transformation of this fungus. This was followed by an adaptation of a plasmid-free CRISPR-Cas9 system coupled with microhomology repair templates. We reproducibly generated stable mutants in the genes leuA and crgA, encoding a 3-isopropylmalate dehydratase and an ubiquitin ligase, respectively. Our new genetic model showed that mutations in the gene pyrF, a key virulence gene in several bacterial and fungal pathogens, correlated with an avirulent phenotype in an immunocompetent murine host. This was reverted by gene complementation, showing the broad possibilities of our methodology.
Collapse
Affiliation(s)
- Carlos Lax
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
| | | | - Carlos Pérez-Arques
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Eusebio Navarro
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
| | - Francisco Esteban Nicolás
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
| | - Victoriano Garre
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
| |
Collapse
|
45
|
2021 in review. Nat Microbiol 2021; 6:1467-1468. [PMID: 34819639 DOI: 10.1038/s41564-021-01016-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
46
|
MohanaSundaram A, Sathanantham ST, Chinchole V, Patil B, Velayutham R. Fostering a deeper understanding of COVID-19-associated mucormycosis - A commentary on "The mucormycosis coinfection in the context of global COVID-19 outbreak: A fatal addition to the pandemic spectrum" (Int. J. Surg. 2021:106031). Int J Surg 2021; 94:106110. [PMID: 34534704 PMCID: PMC8438993 DOI: 10.1016/j.ijsu.2021.106110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/07/2021] [Indexed: 11/06/2022]
Affiliation(s)
- ArunSundar MohanaSundaram
- School of Pharmacy, Sathyabama Institute of Science and Technology, Chennai, Tamilnadu, India School of Pharmaceutical Sciences, Vels Institute of Science, Technology and Advanced Studies (VISTAS), Chennai, Tamilnadu, India Manobal Neuropsychiatric Clinic, Dombivli, Maharashtra, India MannSparsh Neuropsychiatric Hospital, Kalyan, Maharashtra, India Manasa Rehabilitation & De-Addiction Center, Titwala, Maharashtra, India National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, India
| | | | | | | | | |
Collapse
|
47
|
Prakash H, Skiada A, Paul RA, Chakrabarti A, Rudramurthy SM. Connecting the Dots: Interplay of Pathogenic Mechanisms between COVID-19 Disease and Mucormycosis. J Fungi (Basel) 2021; 7:616. [PMID: 34436155 PMCID: PMC8400165 DOI: 10.3390/jof7080616] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/20/2021] [Accepted: 07/26/2021] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease (COVID-19)-associated mucormycosis (CAM) is an emerging threat globally, especially in India. More than 40,000 CAM cases have been reported in India. The emergence of CAM cases in India has been attributed to environmental, host, and iatrogenic factors. Mucorales spore burden has been reported globally; however, their presence is higher in tropical countries such as India, contributing to the emergence of CAM. Before the COVID-19 pandemic, patients with diabetes mellitus, haematological malignancies, solid organ transplants, corticosteroid therapy and neutropenia were more prone to mucormycosis, whereas in COVID-19 patients, virus-induced endothelial dysfunction, hyperglycaemia, and immune dysfunction following corticosteroid use increase the risk of acquiring mucormycosis. The interaction of Mucorales spores with the epithelial cells, followed by endothelial invasion, is a crucial step in the pathogenesis of mucormycosis. Endothelial damage and increased endothelial receptor expression induced by COVID-19 infection may predispose patients to CAM. COVID-19 infection may directly induce hyperglycaemia by damaging beta cells of the pancreas or by corticosteroid therapy, which may contribute to CAM pathogenesis. Iron acquisition from the host, especially in diabetic ketoacidosis (DKA) or deferoxamine therapy, is an important virulence trait of Mucorales. Similarly, the hyperferritinaemia caused by COVID-19 may act as a source of iron for Mucorales growth and invasion. In addition, corticosteroid treatment reduces or abolishes the innate immune functions of phagocytic cells contributing to the pathogenesis of CAM. This review aims to discuss primarily the host and iatrogenic factors shared between COVID-19 and mucormycosis that could explain the emergence of CAM.
Collapse
Affiliation(s)
- Hariprasath Prakash
- Medical Microbiology, Department of Public Health, International Higher School of Medicine, Issyk-Kul Regional Campus, Cholpon-Ata 722125, Kyrgyzstan;
| | - Anna Skiada
- First Department of Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Raees Ahmad Paul
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India; (R.A.P.); (A.C.)
| | - Arunaloke Chakrabarti
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India; (R.A.P.); (A.C.)
| | - Shivaprakash Mandya Rudramurthy
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India; (R.A.P.); (A.C.)
| |
Collapse
|
48
|
Papon N, Naglik JR, Hube B, Goldman GH. Fungal pathogenesis: A new venom. Curr Biol 2021; 31:R391-R394. [PMID: 33905698 DOI: 10.1016/j.cub.2021.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The pathogenesis of life-threatening infections caused by emerging fungal pathogens remains largely unexplored. A new study provides unprecedented evidence for the pivotal role of a new ricin-like protein toxin, named mucoricin, in causing organ necrosis and mortality in Mucorales infections.
Collapse
Affiliation(s)
- Nicolas Papon
- Host-Pathogen Interaction Study Group (GEIHP, EA 3142), UNIV Angers, UNIV Brest, F-49933 Angers, France; Federative Structure of Research, Cellular Interactions and Therapeutic Applications, SFR 4208 ICAT, UNIV Angers, F-49933 Angers, France
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London SE1 1UL, UK
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knoell Institute, Jena 07745, Germany; Institute of Microbiology, Friedrich Schiller University, Jena 07743, Germany
| | - Gustavo H Goldman
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14040-903, Brazil.
| |
Collapse
|
49
|
Cánovas-Márquez JT, Navarro-Mendoza MI, Pérez-Arques C, Lax C, Tahiri G, Pérez-Ruiz JA, Lorenzo-Gutiérrez D, Calo S, López-García S, Navarro E, Nicolás FE, Garre V, Murcia L. Role of the Non-Canonical RNAi Pathway in the Antifungal Resistance and Virulence of Mucorales. Genes (Basel) 2021; 12:genes12040586. [PMID: 33920552 PMCID: PMC8072676 DOI: 10.3390/genes12040586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/12/2021] [Accepted: 04/14/2021] [Indexed: 12/19/2022] Open
Abstract
Mucorales are the causal agents for the lethal disease known as mucormycosis. Mortality rates of mucormycosis can reach up to 90%, due to the mucoralean antifungal drug resistance and the lack of effective therapies. A concerning urgency among the medical and scientific community claims to find targets for the development of new treatments. Here, we reviewed different studies describing the role and machinery of a novel non-canonical RNAi pathway (NCRIP) only conserved in Mucorales. Its non-canonical features are the independence of Dicer and Argonaute proteins. Conversely, NCRIP relies on RNA-dependent RNA Polymerases (RdRP) and an atypical ribonuclease III (RNase III). NCRIP regulates the expression of mRNAs by degrading them in a specific manner. Its mechanism binds dsRNA but only cuts ssRNA. NCRIP exhibits a diversity of functional roles. It represses the epimutational pathway and the lack of NCRIP increases the generation of drug resistant strains. NCRIP also regulates the control of retrotransposons expression, playing an essential role in genome stability. Finally, NCRIP regulates the response during phagocytosis, affecting the multifactorial process of virulence. These critical NCRIP roles in virulence and antifungal drug resistance, along with its exclusive presence in Mucorales, mark this pathway as a promising target to fight against mucormycosis.
Collapse
Affiliation(s)
- José Tomás Cánovas-Márquez
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain; (J.T.C.-M.); (C.L.); (G.T.); (J.A.P.-R.); (D.L.-G.); (S.L.-G.); (E.N.); (F.E.N.); (V.G.)
| | - María Isabel Navarro-Mendoza
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA; (M.I.N.-M.); (C.P.-A.)
| | - Carlos Pérez-Arques
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA; (M.I.N.-M.); (C.P.-A.)
| | - Carlos Lax
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain; (J.T.C.-M.); (C.L.); (G.T.); (J.A.P.-R.); (D.L.-G.); (S.L.-G.); (E.N.); (F.E.N.); (V.G.)
| | - Ghizlane Tahiri
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain; (J.T.C.-M.); (C.L.); (G.T.); (J.A.P.-R.); (D.L.-G.); (S.L.-G.); (E.N.); (F.E.N.); (V.G.)
| | - José Antonio Pérez-Ruiz
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain; (J.T.C.-M.); (C.L.); (G.T.); (J.A.P.-R.); (D.L.-G.); (S.L.-G.); (E.N.); (F.E.N.); (V.G.)
| | - Damaris Lorenzo-Gutiérrez
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain; (J.T.C.-M.); (C.L.); (G.T.); (J.A.P.-R.); (D.L.-G.); (S.L.-G.); (E.N.); (F.E.N.); (V.G.)
| | - Silvia Calo
- School of Natural and Exact Sciences, Pontificia Universidad Católica Madre y Maestra, Santiago de los Caballeros 51033, Dominican Republic;
| | - Sergio López-García
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain; (J.T.C.-M.); (C.L.); (G.T.); (J.A.P.-R.); (D.L.-G.); (S.L.-G.); (E.N.); (F.E.N.); (V.G.)
| | - Eusebio Navarro
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain; (J.T.C.-M.); (C.L.); (G.T.); (J.A.P.-R.); (D.L.-G.); (S.L.-G.); (E.N.); (F.E.N.); (V.G.)
| | - Francisco Esteban Nicolás
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain; (J.T.C.-M.); (C.L.); (G.T.); (J.A.P.-R.); (D.L.-G.); (S.L.-G.); (E.N.); (F.E.N.); (V.G.)
| | - Victoriano Garre
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain; (J.T.C.-M.); (C.L.); (G.T.); (J.A.P.-R.); (D.L.-G.); (S.L.-G.); (E.N.); (F.E.N.); (V.G.)
| | - Laura Murcia
- Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain; (J.T.C.-M.); (C.L.); (G.T.); (J.A.P.-R.); (D.L.-G.); (S.L.-G.); (E.N.); (F.E.N.); (V.G.)
- Correspondence:
| |
Collapse
|
50
|
Brown R, Priest E, Naglik JR, Richardson JP. Fungal Toxins and Host Immune Responses. Front Microbiol 2021; 12:643639. [PMID: 33927703 PMCID: PMC8076518 DOI: 10.3389/fmicb.2021.643639] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/08/2021] [Indexed: 12/11/2022] Open
Abstract
Fungi are ubiquitous organisms that thrive in diverse natural environments including soils, plants, animals, and the human body. In response to warmth, humidity, and moisture, certain fungi which grow on crops and harvested foodstuffs can produce mycotoxins; secondary metabolites which when ingested have a deleterious impact on health. Ongoing research indicates that some mycotoxins and, more recently, peptide toxins are also produced during active fungal infection in humans and experimental models. A combination of innate and adaptive immune recognition allows the host to eliminate invading pathogens from the body. However, imbalances in immune homeostasis often facilitate microbial infection. Despite the wide-ranging effects of fungal toxins on health, our understanding of toxin-mediated modulation of immune responses is incomplete. This review will explore the current understanding of fungal toxins and how they contribute to the modulation of host immunity.
Collapse
Affiliation(s)
| | | | | | - Jonathan P. Richardson
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, United Kingdom
| |
Collapse
|