1
|
Cornely OA, Sprute R, Bassetti M, Chen SCA, Groll AH, Kurzai O, Lass-Flörl C, Ostrosky-Zeichner L, Rautemaa-Richardson R, Revathi G, Santolaya ME, White PL, Alastruey-Izquierdo A, Arendrup MC, Baddley J, Barac A, Ben-Ami R, Brink AJ, Grothe JH, Guinea J, Hagen F, Hochhegger B, Hoenigl M, Husain S, Jabeen K, Jensen HE, Kanj SS, Koehler P, Lehrnbecher T, Lewis RE, Meis JF, Nguyen MH, Pana ZD, Rath PM, Reinhold I, Seidel D, Takazono T, Vinh DC, Zhang SX, Afeltra J, Al-Hatmi AMS, Arastehfar A, Arikan-Akdagli S, Bongomin F, Carlesse F, Chayakulkeeree M, Chai LYA, Chamani-Tabriz L, Chiller T, Chowdhary A, Clancy CJ, Colombo AL, Cortegiani A, Corzo Leon DE, Drgona L, Dudakova A, Farooqi J, Gago S, Ilkit M, Jenks JD, Klimko N, Krause R, Kumar A, Lagrou K, Lionakis MS, Lmimouni BE, Mansour MK, Meletiadis J, Mellinghoff SC, Mer M, Mikulska M, Montravers P, Neoh CF, Ozenci V, Pagano L, Pappas P, Patterson TF, Puerta-Alcalde P, Rahimli L, Rahn S, Roilides E, Rotstein C, Ruegamer T, Sabino R, Salmanton-García J, Schwartz IS, Segal E, Sidharthan N, Singhal T, Sinko J, Soman R, Spec A, Steinmann J, Stemler J, Taj-Aldeen SJ, Talento AF, Thompson GR, Toebben C, Villanueva-Lozano H, Wahyuningsih R, Weinbergerová B, Wiederhold N, Willinger B, Woo PCY, Zhu LP. Global guideline for the diagnosis and management of candidiasis: an initiative of the ECMM in cooperation with ISHAM and ASM. THE LANCET. INFECTIOUS DISEASES 2025:S1473-3099(24)00749-7. [PMID: 39956121 DOI: 10.1016/s1473-3099(24)00749-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/29/2024] [Accepted: 11/08/2024] [Indexed: 02/18/2025]
Abstract
Candida species are the predominant cause of fungal infections in patients treated in hospital, contributing substantially to morbidity and mortality. Candidaemia and other forms of invasive candidiasis primarily affect patients who are immunocompromised or critically ill. In contrast, mucocutaneous forms of candidiasis, such as oral thrush and vulvovaginal candidiasis, can occur in otherwise healthy individuals. Although mucocutaneous candidiasis is generally not life-threatening, it can cause considerable discomfort, recurrent infections, and complications, particularly in patients with underlying conditions such as diabetes or in those taking immunosuppressive therapies. The rise of difficult-to-treat Candida infections is driven by new host factors and antifungal resistance. Pathogens, such as Candida auris (Candidozyma auris) and fluconazole-resistant Candida parapsilosis, pose serious global health risks. Recent taxonomic revisions have reclassified several Candida spp, potentially causing confusion in clinical practice. Current management guidelines are limited in scope, with poor coverage of emerging pathogens and new treatment options. In this Review, we provide updated recommendations for managing Candida infections, with detailed evidence summaries available in the appendix.
Collapse
Affiliation(s)
- Oliver A Cornely
- Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), University of Cologne, Cologne, Germany; Department I of Internal Medicine, European Confederation for Medical Mycology (ECMM) Excellence Center, University of Cologne, Cologne, Germany; German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Cologne, Germany; Clinical Trials Centre Cologne (ZKS Köln), Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany.
| | - Rosanne Sprute
- Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), University of Cologne, Cologne, Germany; Department I of Internal Medicine, European Confederation for Medical Mycology (ECMM) Excellence Center, University of Cologne, Cologne, Germany; German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Cologne, Germany
| | - Matteo Bassetti
- Hospital Policlinico San Martino-IRCCS and Department of Health Science, University of Genoa, Genoa, Italy
| | - Sharon C-A Chen
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research, New South Wales Health Pathology, Westmead Hospital, Sydney, NSW, Australia; Department of Infectious Diseases, Westmead Hospital, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Andreas H Groll
- Infectious Disease Research Program, Center for Bone Marrow Transplantation and Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, University of Münster, Münster, Germany
| | - Oliver Kurzai
- National Reference Center for Invasive Fungal Infections, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knoell-Institute, Jena, Germany; Institute for Hygiene and Microbiology, University of Wuerzburg, Wuerzburg, Germany
| | - Cornelia Lass-Flörl
- Institute for Hygiene and Medical Microbiology, ECMM Excellence Center, Medical University of Innsbruck, Innsbruck, Austria
| | - Luis Ostrosky-Zeichner
- Division of Infectious Diseases, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Riina Rautemaa-Richardson
- Mycology Reference Centre Manchester, ECMM Excellence Center, Department of Infectious Diseases, Manchester Academic Health Science Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK; Division of Evolution, Infection, and Genomics, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, UK
| | - Gunturu Revathi
- Clinical and Diagnostic Microbiology Section, Department of Pathology, Medical College, East Africa, Aga Khan University, Nairobi, Kenya
| | - Maria E Santolaya
- Department of Pediatrics, Infectious Diseases Unit, Hospital Dr Luis Calvo Mackenna, Universidad de Chile, Santiago, Chile
| | - P Lewis White
- Public Health Wales Microbiology Cardiff, University Hospital of Wales, Cardiff, UK; Cardiff University Centre for Trials Research, University Hospital of Wales, Cardiff, UK
| | - Ana Alastruey-Izquierdo
- Center for Biomedical Research in Network in Infectious Diseases, Instituto de Salud Carlos III, Madrid, Spain; Mycology Reference Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III, Majadahonda, Spain
| | - Maiken C Arendrup
- Unit for Mycology, Statens Serum Institut, Copenhagen, Denmark; Department of Clinical Microbiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - John Baddley
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Aleksandra Barac
- Clinic for Infectious and Tropical Diseases, Faculty of Medicine, University Clinical Center of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ronen Ben-Ami
- Department of Infectious Diseases, School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Adrian J Brink
- Division of Medical Microbiology, Faculty of Health Sciences, National Health Laboratory Service, University of Cape Town, Cape Town South Africa; Groote Schuur Hospital, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Jan H Grothe
- Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), University of Cologne, Cologne, Germany; Department I of Internal Medicine, European Confederation for Medical Mycology (ECMM) Excellence Center, University of Cologne, Cologne, Germany; German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Cologne, Germany
| | - Jesus Guinea
- Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, Madrid, Spain; Faculty of Health Sciences, HM Hospitals, Universidad Camilo José Cela, Madrid, Spain
| | - Ferry Hagen
- Department of Medical Mycology, Westerdijk Fungal Biodiversity Institute, Utrecht, Netherlands; Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands; Institute for Biodiversity and Ecosystem Dynamics, University of Amsterdam, Amsterdam, Netherlands
| | - Bruno Hochhegger
- Department of Radiology, University of Florida, Gainesville, FL, USA
| | - Martin Hoenigl
- BioTechMed, Graz, Austria; Division of Infectious Diseases, Translational Medical Mycology Research Unit, ECMM Excellence Center, Medical University of Graz, Graz, Austria
| | - Shahid Husain
- Division of Infectious Diseases, Ajmera Transplant Center, Antimicrobial Stewardship Program University Health Network, University of Toronto, Toronto, ON, Canada
| | - Kauser Jabeen
- Department of Pathology and Laboratory Medicine, Aga Khan University, Karachi, Pakistan
| | - Henrik E Jensen
- Pathology, Section for Pathobiological Sciences, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Souha S Kanj
- Department of Internal Medicine, Division of Infectious Diseases, American University of Beirut Medical Center, Beirut, Lebanon; Center for Infectious Diseases Research, Faculty of Medicine and University Hospital, American University of Beirut Medical Center, Beirut, Lebanon; Department of Internal Medicine, Division of Infectious Diseases, Faculty of Medicine and University Hospital, Duke University Medical Center, Durham, NC, USA
| | - Philipp Koehler
- Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), University of Cologne, Cologne, Germany; Department I of Internal Medicine, European Confederation for Medical Mycology (ECMM) Excellence Center, University of Cologne, Cologne, Germany; German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Cologne, Germany
| | - Thomas Lehrnbecher
- Department of Pediatrics, Division of Hematology, Oncology, and Hemostaseology, Goethe University Frankfurt, Frankfurt, Germany
| | - Russell E Lewis
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Jacques F Meis
- Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center of Expertise for Mycology, Radboud University Medical Center and Canisius-Wilhelmina Hospital, Nijmegen, Netherlands
| | - M Hong Nguyen
- University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zoi D Pana
- Department of Basic and Clinical Studies, University of Nicosia Medical School, Nicosia, Cyprus
| | - Peter-Michael Rath
- Institute for Medical Microbiology, ECMM Excellence Center, University Medicine Essen, University Duisburg-Essen, Essen, Germany
| | - Ilana Reinhold
- Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), University of Cologne, Cologne, Germany; Department I of Internal Medicine, European Confederation for Medical Mycology (ECMM) Excellence Center, University of Cologne, Cologne, Germany
| | - Danila Seidel
- Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), University of Cologne, Cologne, Germany; Department I of Internal Medicine, European Confederation for Medical Mycology (ECMM) Excellence Center, University of Cologne, Cologne, Germany
| | - Takahiro Takazono
- Department of Infectious Diseases, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan; Department of Respiratory Medicine, Nagasaki University Hospital, Nagasaki, Japan
| | - Donald C Vinh
- Centre of Excellence for Genetic Research in Infection and Immunity, Research Institute of the McGill University Health Centre, Montreal, QC, Canada; Division of Infectious Diseases, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada; Division of Medical Microbiology, OPTILAB, Department of Laboratory Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Sean X Zhang
- Microbiology Laboratory, Johns Hopkins Hospital, Baltimore, MD, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Javier Afeltra
- Parasitology and Mycology Unit, Diagnosis and Treatment Department, JM Ramos Mejia Hospital, Department of Immunology, Parasitology and Microbiology, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Abdullah M S Al-Hatmi
- Microbiology Research Laboratory, Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Amir Arastehfar
- Department of Medicine, Harvard Medical School, Boston, MA, USA; Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Sevtap Arikan-Akdagli
- Mycology Unit, Department of Medical Microbiology, Faculty of Medicine, Hacettepe University, Ankara, Türkiye
| | - Felix Bongomin
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Gulu University, Gulu, Uganda
| | - Fabianne Carlesse
- Pediatric Oncology Institute, GRAACC, Federal University of São Paulo, São Paulo, Brazil; Infectious Diseases, Pediatric Department, Federal University of São Paulo, São Paulo, Brazil
| | - Methee Chayakulkeeree
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Louis Y A Chai
- Division of Infectious Diseases, Department of Medicine, National University Health System, Singapore
| | | | - Tom Chiller
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Anuradha Chowdhary
- Medical Mycology Unit, Department of Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India; National Reference Laboratory for Antimicrobial Resistance in Fungal Pathogens, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Cornelius J Clancy
- Infectious Diseases Division, University of Pittsburgh, Pittsburgh, PA, USA
| | - Arnaldo L Colombo
- Department of Medicine, Federal University of São Paulo, São Paulo, Brazil; Antimicrobial Resistance Institute of São Paulo, São Paulo, Brazil
| | - Andrea Cortegiani
- Department of Anaesthesia, Intensive Care, and Emergency, University Hospital Policlinico Paolo Giaccone, Palermo, Italy; Department of Precision Medicine in Medical, Surgical, and Critical Care, University of Palermo, Palermo, Italy
| | - Dora E Corzo Leon
- Medical Research Council Centre for Medical Mycology, ECMM Excellence Center, University of Exeter, Exeter, UK
| | - Lubos Drgona
- Department of Oncohematology, National Cancer Institute, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Anna Dudakova
- Institute for Medical Microbiology, Immunology, and Hygiene, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Joveria Farooqi
- Department of Pathology and Laboratory Medicine, Aga Khan University, Karachi, Pakistan
| | - Sara Gago
- Manchester Fungal Infection Group, School of Biological Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, UK
| | - Macit Ilkit
- Division of Mycology, Department of Microbiology, Faculty of Medicine, University of Çukurova, Adana, Türkiye
| | - Jeffrey D Jenks
- Division of Infectious Diseases, Department of Medicine, Duke University, Durham, NC, USA; Durham County Department of Public Health, Durham, NC, USA
| | - Nikolai Klimko
- Department of Clinical Mycology, Allergology, and Immunology, Northwestern State Medical University named after I I Mechnikov, St Petersburg, Russia
| | - Robert Krause
- BioTechMed, Graz, Austria; Division of Infectious Diseases, Department of Internal Medicine, Medical University of Graz, Austria
| | - Anil Kumar
- Department of Microbiology, Amrita Institute of Medical Sciences, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Katrien Lagrou
- Department of Laboratory Medicine and National Reference Center for Mycosis, ECMM Excellence Center, University Hospitals Leuven, Leuven, Belgium; Laboratory of Clinical Microbiology, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Badre E Lmimouni
- Department of Parasitology and Medical Mycology, Military Teaching Hospital Mohammed the fifth, Faculty of Medicine and Pharmacy, University Mohamed the fifth, Rabat, Morocco
| | - Michael K Mansour
- Department of Medicine, Harvard Medical School, Boston, MA, USA; Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Joseph Meletiadis
- Clinical Microbiology Laboratory, Attikon University General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Sibylle C Mellinghoff
- Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), University of Cologne, Cologne, Germany; Department I of Internal Medicine, European Confederation for Medical Mycology (ECMM) Excellence Center, University of Cologne, Cologne, Germany; German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Cologne, Germany
| | - Mervyn Mer
- Department of Medicine, Divisions of Critical Care and Pulmonology, Charlotte Maxeke Johannesburg Academic Hospital and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Malgorzata Mikulska
- Hospital Policlinico San Martino-IRCCS and Department of Health Science, University of Genoa, Genoa, Italy; Division of Infectious Diseases, Department of Health Sciences, University of Genova, Genova, Italy
| | - Philippe Montravers
- Department of Anaesthesiology and Critical Care Medicine, CHU-Bichat Claude Bernard, AP-HP Nord, Assistance Publique-Hôpitaux de Paris, Paris, France; Physiopathology and Epidemiology of Respiratory Diseases, French Institute of Health and Medical Research (INSERM), U1152, University Paris-Cité, Paris, France; UFR Médecine Paris Cité, University Paris-Cité, Paris, France
| | - Chin Fen Neoh
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Volkan Ozenci
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Livio Pagano
- Department of Geriatric Hematology, Fondazione Policlinico Universitario A Gemelli-IRCCS, Università del Sacro Cuore, Rome, Italy
| | - Peter Pappas
- Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Thomas F Patterson
- Department of Medicine, Division of Infectious Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Pedro Puerta-Alcalde
- Department of Infectious Diseases, Hospital Clinic of Barcelona-IDIBAPS, Barcelona, Spain; Department of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Laman Rahimli
- Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), University of Cologne, Cologne, Germany; Department I of Internal Medicine, European Confederation for Medical Mycology (ECMM) Excellence Center, University of Cologne, Cologne, Germany
| | - Sebastian Rahn
- Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), University of Cologne, Cologne, Germany; Department I of Internal Medicine, European Confederation for Medical Mycology (ECMM) Excellence Center, University of Cologne, Cologne, Germany; German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Cologne, Germany
| | - Emmanuel Roilides
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece; Hippokration General Hospital, Thessaloniki, Greece; Infectious Diseases Unit, 3rd Department of Pediatrics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Coleman Rotstein
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Tamara Ruegamer
- Institute for Medical Microbiology, Immunology, and Hygiene, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Raquel Sabino
- Department of Pharmacy, Pharmacology and Health Technologies, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal; Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal; Laboratório Associado TERRA-Laboratório para o Uso Sustentável da Terra e dos Serviços dos Ecossistemas, Instituto Superior de Agronomia, Lisbon, Portugal
| | - Jon Salmanton-García
- Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), University of Cologne, Cologne, Germany; Department I of Internal Medicine, European Confederation for Medical Mycology (ECMM) Excellence Center, University of Cologne, Cologne, Germany; German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Cologne, Germany
| | - Ilan S Schwartz
- Department of Internal Medicine, Division of Infectious Diseases, Faculty of Medicine and University Hospital, Duke University Medical Center, Durham, NC, USA
| | - Esther Segal
- Department of Clinical Microbiology and Immunology, School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Tanu Singhal
- Consultant Paediatrics and Infectious Disease, Kokilaben Dhirubhai Ambani Hospital and Medical Research Institute, Mumbai, India
| | - Janos Sinko
- South-Pest Central Hospital-National Institute of Hematology and Infectology, Budapest, Hungary
| | - Rajeev Soman
- Department of Infectious Diseases, Jupiter Hospital, Pune, India
| | - Andrej Spec
- Division of Infectious Diseases, Washington University School of Medicine, ECMM Excellence Center, St Louis, MO, USA
| | - Joerg Steinmann
- Institute of Clinical Microbiology, Infectious Diseases and Infection Control, Klinikum Nuremberg, Paracelsus Medical University, Nuremberg, Germany; Institute of Medical Microbiology, University Hospital Essen, Essen, Germany
| | - Jannik Stemler
- Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), University of Cologne, Cologne, Germany; Department I of Internal Medicine, European Confederation for Medical Mycology (ECMM) Excellence Center, University of Cologne, Cologne, Germany; German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Cologne, Germany
| | - Saad J Taj-Aldeen
- Department of Biology, College of Science, University of Babylon, Hilla, Iraq; Microbiology Laboratory, Department of Laboratory Medicine and Pathology, Hamad Medical Corporation, Doha, Qatar
| | - Alida Fe Talento
- Department of Clinical Microbiology, ECMM Excellence Center, Trinity College Dublin, Dublin, Ireland; Department of Microbiology, Children's Health Ireland at Temple Street, Dublin, Ireland; Department of Microbiology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - George R Thompson
- Department of Internal Medicine, Division of Infectious Diseases, University of California Davis Medical Center, Sacramento, CA, USA
| | - Christina Toebben
- Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), University of Cologne, Cologne, Germany; Department I of Internal Medicine, European Confederation for Medical Mycology (ECMM) Excellence Center, University of Cologne, Cologne, Germany; German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Cologne, Germany
| | - Hiram Villanueva-Lozano
- Division of Infectious Diseases, Department of Internal Medicine, Hospital Regional Monterrey, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, Monterrey, Mexico
| | | | - Barbora Weinbergerová
- Department of Internal Medicine, Hematology, and Oncology, University Hospital Brno, Masaryk University, Brno, Czech Republic
| | - Nathan Wiederhold
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Birgit Willinger
- Department for Laboratory Medicine, Division of Clinical Microbiology, ECMM Excellence Center, Medical University of Vienna, Vienna, Austria
| | - Patrick C Y Woo
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan; Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong Special Administrative Region, China; The iEGG and Animal Biotechnology Research Center, National Chung Hsing University, Taichung, Taiwan
| | - Li-Ping Zhu
- Department of Infectious Diseases, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Epelbaum O, Marinelli T, Haydour QS, Pennington KM, Evans SE, Carmona EM, Husain S, Knox KS, Jarrett BJ, Azoulay E, Hope WW, Meyer-Zilla A, Murad MH, Limper AH, Hage CA. Treatment of Invasive Pulmonary Aspergillosis and Preventive and Empirical Therapy for Invasive Candidiasis in Adult Pulmonary and Critical Care Patients. An Official American Thoracic Society Clinical Practice Guideline. Am J Respir Crit Care Med 2024; 211:34-53. [PMID: 39556361 PMCID: PMC11755356 DOI: 10.1164/rccm.202410-2045st] [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: 10/22/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND The incidence of invasive fungal infections is increasing in immune-competent and immune-compromised patients. An examination of the recent literature related to the treatment of fungal infections was performed to address two clinical questions. First, in patients with proven or probable invasive pulmonary aspergillosis, should combination therapy with a mold-active triazole plus echinocandin be administered vs. mold-active triazole monotherapy? Second, in critically ill patients at risk for invasive candidiasis who are non-neutropenic and are not transplant recipients, should systemic antifungal agents be administered either as prophylaxis or as empiric therapy? METHODS A multidisciplinary panel reviewed the available data concerning the two questions. The evidence was evaluated, and recommendations were generated using the Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) approach. RESULTS A conditional recommendation was made for patients with proven or probable invasive pulmonary aspergillosis to receive either initial combination therapy with a mold-active triazole plus an echinocandin or initial mold-active triazole monotherapy based on low-quality evidence. Further, a conditional weak recommendation was made against routine administration of prophylactic or empiric antifungal agents targeting Candida species for critically ill patients without neutropenia or a history of transplant based on low-quality evidence. CONCLUSIONS The recommendations presented in these Guidelines are the result of an analysis of currently available evidence. Additional research and new clinical data will prompt an update in the future.
Collapse
Affiliation(s)
- Oleg Epelbaum
- Westchester Medical Center, Pulmonary and Critical Care Medicine, Valhalla, New York, United States
| | - Tina Marinelli
- Royal Prince Alfred Hospital, Infectious Diseases, Sydney, Australia
| | | | - Kelly M Pennington
- Mayo Clinic, Pulmonary and Critical Care , Rochester, Minnesota, United States
| | - Scott E Evans
- University of Texas-M.D. Anderson Cancer Center, Pulmonary Medicine, Houston, Texas, United States
| | - Eva M Carmona
- Mayo Clinic and Foundation, Pulmonary and Critical Care Medicine, Rochester, Minnesota, United States
| | - Shahid Husain
- University Health Network , University of Toronto, Depatment of Medicine, Multiorgan Transplantation Institute, Toronto, Ontario, Canada
| | - Kenneth S Knox
- University of Arizona Medical Center - University Campus, Medicine, Tucson, Arizona, United States
| | | | | | - William W Hope
- University of Liverpool, Liverpool, United Kingdom of Great Britain and Northern Ireland
| | | | - M Hassan Murad
- Mayo Clinic, Evidence-Based Practice Center, Rochester, Minnesota, United States
| | | | - Chadi A Hage
- University of Pittsburgh, Pittsburgh, Pennsylvania, United States;
| |
Collapse
|
3
|
Nyumura Y, Tsuboi K, Suzuki T, Kajimoto T, Tanishima Y, Yano F, Eto K. Pathophysiology and surgical outcomes of patients with fungal peritonitis from upper gastrointestinal tract perforation: a retrospective study. Surg Today 2024; 54:1345-1352. [PMID: 38691220 DOI: 10.1007/s00595-024-02851-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 03/26/2024] [Indexed: 05/03/2024]
Abstract
PURPOSE To compare the pathophysiology and surgical outcomes of emergency surgery for upper gastrointestinal tract perforation with and without fungal peritonitis and identify the risk factors for fungal peritonitis. METHODS The subjects of this retrospective study were patients with upper gastrointestinal perforation and peritonitis who underwent emergency surgery at a single medical center in Japan. The patients were allocated to two groups according to the presence or absence of fungal peritonitis: group F and group N, respectively. RESULTS At the time of surgery, ascitic fluid culture or serum β-D glucan levels were available for 54 patients: 29 from group F and 25 from group N, respectively. The stomach was perforated in 14 patients (25.9%) and the duodenum was perforated in 40 patients (74.1%). Group F had a higher proportion of patients with low preoperative prognostic nutritional index scores (≤ 40) and C-reactive protein levels and a higher postoperative complication rate. The time to initiate food intake and the postoperative hospital stay were also significantly longer in group F. Multivariate analysis identified that the perforation site of the stomach was a risk factor for fungal peritonitis. CONCLUSION Patients with fungal peritonitis from upper gastrointestinal tract perforation had higher postoperative complication rates, delayed postoperative recovery, and a longer hospital stay. Gastric perforation was a risk factor for fungal peritonitis.
Collapse
Affiliation(s)
- Yuya Nyumura
- Department of Surgery, Fuji City General Hospital, 50, Takashima-Cho, Fuji-Shi, Shizuoka, 417-8567, Japan.
- Department of Gastrointestinal Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan.
| | - Kazuto Tsuboi
- Department of Surgery, Fuji City General Hospital, 50, Takashima-Cho, Fuji-Shi, Shizuoka, 417-8567, Japan
| | - Toshimasa Suzuki
- Department of Surgery, Fuji City General Hospital, 50, Takashima-Cho, Fuji-Shi, Shizuoka, 417-8567, Japan
| | - Tetsuya Kajimoto
- Department of Surgery, Fuji City General Hospital, 50, Takashima-Cho, Fuji-Shi, Shizuoka, 417-8567, Japan
| | - Yuichiro Tanishima
- Department of Gastrointestinal Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Fumiaki Yano
- Department of Gastrointestinal Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Ken Eto
- Department of Gastrointestinal Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| |
Collapse
|
4
|
Fandilolu P, Kumar C, Palia D, Idicula-Thomas S. Investigating role of positively selected genes and mutation sites of ERG11 in drug resistance of Candida albicans. Arch Microbiol 2024; 206:437. [PMID: 39422772 DOI: 10.1007/s00203-024-04159-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024]
Abstract
The steep increase in acquired drug resistance in Candida isolates has posed a great challenge in the clinical management of candidiasis globally. Information of genes and codon sites that are positively selected during evolution can provide insights into the mechanisms driving antifungal resistance in Candida. This study aimed to create a manually curated list of genes of Candida spp. reported to be associated with antifungal resistance in literature, and further investigate the structure-function implications of positively selected genes and mutation sites. Sequence analysis of antifungal drug resistance associated gene sequences from various species and strains of Candida revealed that ERG11 and MRR1 of C. albicans were positively selected during evolution. Four sites in ERG11 and two sites in MRR1 of C. albicans were positively selected and associated with drug resistance. These four sites (132, 405, 450, and 464) of ERG11 are predictive markers for azole resistance and have evolved over time. A well-characterized crystal structure of sterol-14-α-demethylase (CYP51) encoded by ERG11 is available in PDB. Therefore, the stability of CYP51 in complex with fluconazole was evaluated using MD simulations and molecular docking studies for two mutations (Y132F and Y132H) reported to be associated with azole resistance in literature. These mutations induced high flexibility in functional motifs of CYP51. It was also observed that residues such as I304, G308, and I379 of CYP51 play a critical role in fluconazole binding affinity. The insights gained from this study can further guide drug design strategies addressing antimicrobial resistance.
Collapse
Affiliation(s)
- Prayagraj Fandilolu
- Biomedical Informatics Centre, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, 400012, India
| | - Chandan Kumar
- Biomedical Informatics Centre, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, 400012, India
| | - Dushyant Palia
- Biomedical Informatics Centre, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, 400012, India
| | - Susan Idicula-Thomas
- Biomedical Informatics Centre, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, 400012, India.
| |
Collapse
|
5
|
Xu ZC, Ma XR, Zhang LJ, Chen HT, Qing DM, Li RT, Ye RR, Wang RR. Antifungal activity of ruthenium (II) complex combined with fluconazole against drug-resistant Candida albicans in vitro and its anti-invasive infection in vivo. J Inorg Biochem 2024; 255:112522. [PMID: 38522215 DOI: 10.1016/j.jinorgbio.2024.112522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/29/2024] [Accepted: 03/08/2024] [Indexed: 03/26/2024]
Abstract
With the abuse of antibiotics and azoles, drug-resistant Candida albicans infections have increased sharply and are spreading rapidly, thereby significantly reducing the antifungal efficacy of existing therapeutics. Several patients die of fungal infections every year. Therefore, there is an urgent requirement to develop new drugs. Accordingly, we synthesized a series of polypyridyl ruthenium (II) complexes having the formula [Ru (NN)2 (bpm)] (PF6)2 (N-N = 2,2'-bipyridine) (bpy, in Ru1), 1,10-phenanthroline (phen, in Ru2), 4,7-diphenyl-1,10-phenanthroline (DIP, in Ru3) (bpm = 2,2'-bipyrimidine) and studied their antifungal activities. Ru3 alone had no effect on the drug-resistant strains, but Ru3 combined with fluconazole (FLC) exhibited significant antifungal activity on drug-resistant strains. A high-dose combination of Ru3 and FLC exhibited direct fungicidal activity by promoting the accumulation of reactive oxygen species and damaging the cellular structure of C. albicans. Additionally, the combination of Ru3 and FLC demonstrated potent antifungal efficacy in vivo in a mouse model of invasive candidiasis. Moreover, the combination significantly improved the survival state of mice, restored their immune systems, and reduced renal injury. These findings could provide ideas for the development of ruthenium (II) complexes as novel antifungal agents for drug-resistant microbial stains.
Collapse
Affiliation(s)
- Zhi-Chang Xu
- College of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Xiu-Rong Ma
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Li-Juan Zhang
- College of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Hui-Ting Chen
- College of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Ding-Mei Qing
- College of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Rong-Tao Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Rui-Rong Ye
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China.
| | - Rui-Rui Wang
- College of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China.
| |
Collapse
|
6
|
Khichi A, Jakhar R, Dahiya S, Arya J, Dangi M, Chhillar AK. In silico and in vitro evaluation of designed fluconazole analogues as lanosterol 14α-demethylase inhibitors. J Biomol Struct Dyn 2024; 42:4553-4566. [PMID: 37293950 DOI: 10.1080/07391102.2023.2220808] [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: 02/02/2023] [Accepted: 05/29/2023] [Indexed: 06/10/2023]
Abstract
The drugs fighting against aggressive fungal infections are in limited number, therefore, extensive research is obligatory to develop new therapeutic strategies. Fluconazole (FLZ) is a clinically approved drug, but resistant drug against most fungal pathogens, thus it is vital to identify more compounds that can better check the fungal growth. Analogue-based drug designing is a quick and economical way since it has inherent drug-like properties of marketed drugs. This study aims to generate and evaluate analogues of FLZ with better potency against fungal-borne infections. A total of 3307 analogues of FLZ were developed from six scaffold structures. Only 390 compounds passed Lipinski's rule, of which 247 analogues exhibited lower docking scores than FLZ with 5FSA. These inhibitors were further subjected to pharmacokinetics property evaluation and cytotoxicity test and it was found that only 46 analogues were suitable for further evaluation. Based on the molecular docking score of the best two analogues, 6f (-12.7 kcal/mol) and 8f (-12.8 kcal/mol) were selected for molecular dynamics and in-vitro studies. Antifungal activities of both compounds against 4 strains of Candida albicans were evaluated by disc diffusion assay and micro broth dilution assay and Minimum inhibitory concentrations (MICs) for 6f and 8f were observed as 256 µg/ml against 4719, 4918 and 5480 strains but the MIC was extended to 512 µg/ml for strain 3719. Both analogues exhibited low antifungal activities as compared to FLZ (8-16 µg/ml). The interaction of 6f with Mycostatin was also performed using a chequerboard assay that was found additive.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Alka Khichi
- Centre for Bioinformatics, Maharshi Dayanand University, Rohtak, India
| | - Ritu Jakhar
- Centre for Bioinformatics, Maharshi Dayanand University, Rohtak, India
| | - Sweety Dahiya
- Centre for Biotechnology, Maharshi Dayanand University, Rohtak, India
| | - Jasmine Arya
- Centre for Biotechnology, Maharshi Dayanand University, Rohtak, India
| | - Mehak Dangi
- Centre for Bioinformatics, Maharshi Dayanand University, Rohtak, India
| | | |
Collapse
|
7
|
Liu S, Tian H, Ming H, Zhang T, Gao Y, Liu R, Chen L, Yang C, Nice EC, Huang C, Bao J, Gao W, Shi Z. Mitochondrial-Targeted CS@KET/P780 Nanoplatform for Site-Specific Delivery and High-Efficiency Cancer Immunotherapy in Hepatocellular Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308027. [PMID: 38308137 PMCID: PMC11005749 DOI: 10.1002/advs.202308027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/07/2024] [Indexed: 02/04/2024]
Abstract
Hepatocellular carcinoma (HCC) is a form of malignancy with limited curative options available. To improve therapeutic outcomes, it is imperative to develop novel, potent therapeutic modalities. Ketoconazole (KET) has shown excellent therapeutic efficacy against HCC by eliciting apoptosis. However, its limited water solubility hampers its application in clinical treatment. Herein, a mitochondria-targeted chemo-photodynamic nanoplatform, CS@KET/P780 NPs, is designed using a nanoprecipitation strategy by integrating a newly synthesized mitochondria-targeted photosensitizer (P780) and chemotherapeutic agent KET coated with chondroitin sulfate (CS) to amplify HCC therapy. In this nanoplatform, CS confers tumor-targeted and subsequently pH-responsive drug delivery behavior by binding to glycoprotein CD44, leading to the release of P780 and KET. Mechanistically, following laser irradiation, P780 targets and destroys mitochondrial integrity, thus inducing apoptosis through the enhancement of reactive oxygen species (ROS) buildup. Meanwhile, KET-induced apoptosis synergistically enhances the anticancer effect of P780. In addition, tumor cells undergoing apoptosis can trigger immunogenic cell death (ICD) and a longer-term antitumor response by releasing tumor-associated antigens (TAAs) and damage-associated molecular patterns (DAMPs), which together contribute to improved therapeutic outcomes in HCC. Taken together, CS@KET/P780 NPs improve the bioavailability of KET and exhibit excellent therapeutic efficacy against HCC by exerting chemophototherapy and antitumor immunity.
Collapse
Affiliation(s)
- Shanshan Liu
- Clinical Medical CollegeAffiliated Hospital of Chengdu UniversityChengdu UniversityChengdu610106China
- Department of Clinical PharmacySchool of PharmacyZunyi Medical UniversityZunyi563006China
| | - Hailong Tian
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospitaland West China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityCollaborative Innovation Center for BiotherapyChengdu610041China
| | - Hui Ming
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospitaland West China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityCollaborative Innovation Center for BiotherapyChengdu610041China
| | - Tingting Zhang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospitaland West China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityCollaborative Innovation Center for BiotherapyChengdu610041China
| | - Yajie Gao
- The First Affiliated Hospital of Ningbo UniversityNingbo315020China
| | - Ruolan Liu
- School of Basic Medical SciencesChengdu University of Traditional Chinese MedicineChengdu611137China
| | - Lihua Chen
- School of Basic Medical SciencesChengdu University of Traditional Chinese MedicineChengdu611137China
| | - Chen Yang
- School of Basic Medical SciencesChengdu University of Traditional Chinese MedicineChengdu611137China
| | - Edouard C. Nice
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVIC3800Australia
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospitaland West China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityCollaborative Innovation Center for BiotherapyChengdu610041China
| | - Jinku Bao
- College of Life SciencesSichuan UniversityChengdu610064China
| | - Wei Gao
- Clinical Medical CollegeAffiliated Hospital of Chengdu UniversityChengdu UniversityChengdu610106China
- Clinical Genetics LaboratoryAffiliated Hospital & Clinical Medical College of Chengdu UniversityChengdu610081China
| | - Zheng Shi
- Clinical Medical CollegeAffiliated Hospital of Chengdu UniversityChengdu UniversityChengdu610106China
- Department of Clinical PharmacySchool of PharmacyZunyi Medical UniversityZunyi563006China
| |
Collapse
|
8
|
Kurakado S, Matsumoto Y, Eshima S, Sugita T. Antimicrobial Tolerance in Cross-Kingdom Dual-Species Biofilms Formed by Fungi and Bacteria. Med Mycol J 2024; 65:49-57. [PMID: 39218647 DOI: 10.3314/mmj.24.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Candida albicans, the most common pathogenic fungus, can form biofilms on the surface of medical devices and often causes bloodstream infections. Biofilms have a complex structure composed of microorganisms and a surrounding extracellular matrix. Biofilms are difficult to treat because they are resistant to antifungal drugs and the host environment. Nearly one in four patients with candidemia have a polymicrobial infection. These polymicrobial biofilms, especially those comprising cross-kingdom species of fungi and bacteria, can lead to long hospital stays and high mortality rates. This review outlines the unique interactions of dual-species biofilms with Candida albicans and the clinically important bacteria Staphylococcus aureus, Pseudomonas aeruginosa, and Escherichia coli.
Collapse
Affiliation(s)
- Sanae Kurakado
- Department of Microbiology, Meiji Pharmaceutical University
| | | | | | - Takashi Sugita
- Department of Microbiology, Meiji Pharmaceutical University
| |
Collapse
|
9
|
Habighorst K, Sanders JM, Hennessy SA, Goff K, Wan B, Johns M. Identification of Risk Factors for Intra-Abdominal Candidiasis. Surg Infect (Larchmt) 2023; 24:910-915. [PMID: 38011638 DOI: 10.1089/sur.2023.149] [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: 11/29/2023] Open
Abstract
Background: Intra-abdominal candidiasis (IAC) is associated with substantial morbidity and mortality in hospitalized patients. Identifying high-risk populations may facilitate early and selective directed therapy in appropriate patients and avoid unwarranted treatment and any associated adverse effects in those who are low risk. Patients and Methods: This retrospective, case-control study included patients >18 years of age admitted from July 1, 2010 to July 1, 2021 who had a microbiologically confirmed intra-abdominal infection (gastrointestinal culture positive for either a Candida spp. [cases] or bacterial isolate [controls] collected intra-operatively or from a drain placed within 24 hours). Patients receiving peritoneal dialysis treatment or with a peritoneal dialysis catheter in place or treated at an outside hospital were excluded. Multivariable regression was utilized to identify independent risk factors for the development of IAC. Results: Five hundred twenty-three patients were screened, and 250 met inclusion criteria (125 per cohort). Multivariable analysis identified exposure to corticosteroids (odds ratio [OR], 5.79; 95% confidence interval [CI], 2.52-13.32; p < 0.0001), upper gastrointestinal tract surgery (OR, 3.51; 95% CI, 1.25-9.87; p = 0.017), and mechanical ventilation (OR, 3.09; 95% CI 1.5-6.37; p = 0.002) were independently associated with IAC. The area under the receiver operating characteristic (AUROC) and goodness of fit were 0.7813 and p = 0.5024, respectively. Conclusions: Exposure to corticosteroids, upper gastrointestinal tract surgery, and mechanical ventilation are independent risk factors for the development of microbiologically confirmed IAC suggesting these factors may help identify high-risk individuals requiring antifungal therapy.
Collapse
Affiliation(s)
- Kelsey Habighorst
- Department of Pharmacy, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - James M Sanders
- Department of Pharmacy, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Division of Infectious Diseases and Geographic Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sara A Hennessy
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kristina Goff
- Department of Anesthesiology and Pain Management, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Bingchun Wan
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Meagan Johns
- Department of Pharmacy, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
10
|
Lohse MB, Ziv N, Johnson AD. Variation in transcription regulator expression underlies differences in white-opaque switching between the SC5314 reference strain and the majority of Candida albicans clinical isolates. Genetics 2023; 225:iyad162. [PMID: 37811798 PMCID: PMC10627253 DOI: 10.1093/genetics/iyad162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 08/26/2023] [Indexed: 10/10/2023] Open
Abstract
Candida albicans, a normal member of the human microbiome and an opportunistic fungal pathogen, undergoes several morphological transitions. One of these transitions is white-opaque switching, where C. albicans alternates between 2 stable cell types with distinct cellular and colony morphologies, metabolic preferences, mating abilities, and interactions with the innate immune system. White-to-opaque switching is regulated by mating type; it is repressed by the a1/α2 heterodimer in a/α cells, but this repression is lifted in a/a and α/α mating type cells (each of which are missing half of the repressor). The widely used C. albicans reference strain, SC5314, is unusual in that white-opaque switching is completely blocked when the cells are a/α; in contrast, most other C. albicans a/α strains can undergo white-opaque switching at an observable level. In this paper, we uncover the reason for this difference. We show that, in addition to repression by the a1/α2 heterodimer, SC5314 contains a second block to white-opaque switching: 4 transcription regulators of filamentous growth are upregulated in this strain and collectively suppress white-opaque switching. This second block is missing in the majority of clinical strains, and, although they still contain the a1/α2 heterodimer repressor, they exhibit a/α white-opaque switching at an observable level. When both blocks are absent, white-opaque switching occurs at very high levels. This work shows that white-opaque switching remains intact across a broad group of clinical strains, but the precise way it is regulated and therefore the frequency at which it occurs varies from strain to strain.
Collapse
Affiliation(s)
- Matthew B Lohse
- Department of Microbiology and Immunology, University of California - San Francisco, San Francisco, CA 94143, USA
| | - Naomi Ziv
- Department of Microbiology and Immunology, University of California - San Francisco, San Francisco, CA 94143, USA
| | - Alexander D Johnson
- Department of Microbiology and Immunology, University of California - San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
11
|
Arafa SH, Elbanna K, Osman GEH, Abulreesh HH. Candida diagnostic techniques: a review. JOURNAL OF UMM AL-QURA UNIVERSITY FOR APPLIED SCIENCES 2023; 9:360-377. [DOI: 10.1007/s43994-023-00049-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/27/2023] [Indexed: 01/03/2025]
Abstract
AbstractFungal infections (mycoses) represent a major health issue in humans. They have emerged as a global concern for medical professionals by causing high morbidity and mortality. Fungal infections approximately impact one billion individuals per annum and account for 1.6 million deaths. The diagnosis of Candida infections is a challenging task. Laboratory-based Candida species identification techniques (molecular, commercial, and conventional) have been reviewed and summarized. This review aims to discuss the mycoses history, taxonomy, pathogenicity, and virulence characteristics.
Collapse
|
12
|
Mauri C, Consonni A, Briozzo E, Giubbi C, Meroni E, Tonolo S, Luzzaro F. Microbiological Assessment of the FilmArray Blood Culture Identification 2 Panel: Potential Impact in Critically Ill Patients. Antibiotics (Basel) 2023; 12:1247. [PMID: 37627667 PMCID: PMC10451250 DOI: 10.3390/antibiotics12081247] [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: 06/30/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Rapid pathogen detection and characterization from positive blood cultures are crucial in the management of patients with bloodstream infections (BSI) and in achieving their improved outcomes. In this context, the FilmArray Blood Culture Identification (BCID2) panel is an FDA approved molecular test, which can quickly identify different species and resistance determinants, thus making an impact in antimicrobial practice. In this study, we analyzed 136 positive blood cultures collected from septic critically ill patients from April 2021 to March 2023 by using the FilmArray BCID2 panel, and results obtained by fast molecular analysis were compared to those obtained by routine protocols. Overall, the BCID2 panel showed a strong concordance with conventional methods, particularly in the case of monomicrobial samples, whereas some discrepancies were found in 10/32 polymicrobial samples. Of note, this technique allowed us to identify a significant number of yeasts (37/94 samples) and to unravel the presence of several resistance markers, including both Gram-positive and Gram-negative organisms. These findings strongly support the potential use of the BCID2 panel as an adjunct to the conventional microbiology methods for the management of critically ill septic patients, thus accelerating blood pathogen and resistance genes identification, focusing antibiotic therapy, and avoiding inappropriate and excessive use of drugs.
Collapse
Affiliation(s)
- Carola Mauri
- Clinical Microbiology and Virology Unit, “A. Manzoni” Hospital, 23900 Lecco, Italy
| | | | | | | | | | | | | |
Collapse
|
13
|
Kangabam N, Nethravathy V. An overview of opportunistic fungal infections associated with COVID-19. 3 Biotech 2023; 13:231. [PMID: 37309405 PMCID: PMC10257773 DOI: 10.1007/s13205-023-03648-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 05/26/2023] [Indexed: 06/14/2023] Open
Abstract
The COVID-19 survivors and long-term steroid administered patients exhibit a variety of fungal co-infections. The lives of COVID-19 patients and survivors are hampered by fungal species of the genera Candida, Aspergillus, and Mucor. There have been cases of mucormycosis, aspergillosis, and candidiasis in COVID-19 patients. The treatments given to these opportunistic fungal infections include polyene like amphotericin B, azoles including imidazoles like ketoconazole, miconazole, and triazoles like fluconazole, voriconazole, itraconazole, Echinocandin derivatives like- caspofungin, micafungin, immunomodulatory therapy, granulocyte transfusion, etc. A successful recovery and the reduction of fatalities depend on prompt diagnosis and treatment. To reduce mortality, advanced techniques to identify such uncommon infections at a very early stage are necessary. This review's goal is to provide a summary of the systemic and superficial opportunistic fungal infections that the COVID-19 survivors were dealing with, including information on illness incidence, pathogenicity, and treatment.
Collapse
Affiliation(s)
- Navidita Kangabam
- Department of Biotechnology, School of Applied Sciences, REVA University, , Rukmini Knowledge Park, Kattigenehalli, Yelahanka, Bengaluru, 560064 India
| | - V. Nethravathy
- Department of Biotechnology, School of Applied Sciences, REVA University, , Rukmini Knowledge Park, Kattigenehalli, Yelahanka, Bengaluru, 560064 India
| |
Collapse
|
14
|
Elalouf A. Infections after organ transplantation and immune response. Transpl Immunol 2023; 77:101798. [PMID: 36731780 DOI: 10.1016/j.trim.2023.101798] [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: 08/26/2022] [Revised: 01/08/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023]
Abstract
Organ transplantation has provided another chance of survival for end-stage organ failure patients. Yet, transplant rejection is still a main challenging factor. Immunosuppressive drugs have been used to avoid rejection and suppress the immune response against allografts. Thus, immunosuppressants increase the risk of infection in immunocompromised organ transplant recipients. The infection risk reflects the relationship between the nature and severity of immunosuppression and infectious diseases. Furthermore, immunosuppressants show an immunological impact on the genetics of innate and adaptive immune responses. This effect usually reactivates the post-transplant infection in the donor and recipient tissues since T-cell activation has a substantial role in allograft rejection. Meanwhile, different infections have been found to activate the T-cells into CD4+ helper T-cell subset and CD8+ cytotoxic T-lymphocyte that affect the infection and the allograft. Therefore, the best management and preventive strategies of immunosuppression, antimicrobial prophylaxis, and intensive medical care are required for successful organ transplantation. This review addresses the activation of immune responses against different infections in immunocompromised individuals after organ transplantation.
Collapse
Affiliation(s)
- Amir Elalouf
- Bar-Ilan University, Department of Management, Ramat Gan 5290002, Israel.
| |
Collapse
|
15
|
Lim H, Kim JY, Choo S, Lee C, Han BJ, Lim CS, Nam J. Separation and Washing of Candida Cells from White Blood Cells Using Viscoelastic Microfluidics. MICROMACHINES 2023; 14:712. [PMID: 37420947 DOI: 10.3390/mi14040712] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/16/2023] [Accepted: 03/22/2023] [Indexed: 07/09/2023]
Abstract
An early and accurate diagnosis of Candida albicans is critical for the rapid antifungal treatment of candidemia, a mortal bloodstream infection. This study demonstrates viscoelastic microfluidic techniques for continuous separation, concentration, and subsequent washing of Candida cells in the blood. The total sample preparation system contains two-step microfluidic devices: a closed-loop separation and concentration device and a co-flow cell-washing device. To determine the flow conditions of the closed-loop device, such as the flow rate factor, a mixture of 4 and 13 μm particles was used. Candida cells were successfully separated from the white blood cells (WBCs) and concentrated by 74.6-fold in the sample reservoir of the closed-loop system at 800 μL/min with a flow rate factor of 3.3. In addition, the collected Candida cells were washed with washing buffer (deionized water) in the microchannels with an aspect ratio of 2 at a total flow rate of 100 μL/min. Finally, Candida cells at extremely low concentrations (Ct > 35) became detectable after the removal of WBCs, the additional buffer solution in the closed-loop system (Ct = 30.3 ± 1.3), and further removal of blood lysate and washing (Ct = 23.3 ± 1.6).
Collapse
Affiliation(s)
- Hyunjung Lim
- School of Biomedical Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Jae Young Kim
- Research Institute for Skin Image, Korea University College of Medicine, Seoul 08308, Republic of Korea
- Core Research & Development Center, Korea University Ansan Hospital, Ansan 15355, Republic of Korea
| | - Seunghee Choo
- College of Life Sciences and Bio Engineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Changseok Lee
- Department of AI Electrical and Electronic Engineering, Incheon Jaeneung University, Incheon 22573, Republic of Korea
| | - Byoung Joe Han
- Department of AI Electrical and Electronic Engineering, Incheon Jaeneung University, Incheon 22573, Republic of Korea
| | - Chae Seung Lim
- Department of Laboratory Medicine, College of Medicine, Korea University, Seoul 08307, Republic of Korea
| | - Jeonghun Nam
- Department of AI Electrical and Electronic Engineering, Incheon Jaeneung University, Incheon 22573, Republic of Korea
- Artificial Intelligence(AI)-Bio Research Center, Incheon Jaeneung University, Incheon 21987, Republic of Korea
| |
Collapse
|
16
|
Villavicencio A, Ruiz EF, Fernandez O, Fernandez A, Gonzales‐Zamora JA. Such a fun-guy! Is this Candida or Histoplasma? Clin Case Rep 2023; 11:e7144. [PMID: 36992668 PMCID: PMC10040493 DOI: 10.1002/ccr3.7144] [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: 11/15/2022] [Revised: 02/13/2023] [Accepted: 03/14/2023] [Indexed: 03/29/2023] Open
Abstract
Histoplasmosis is a neglected mycosis with high mortality in immunocompromised individuals. The diagnosis can be delayed due to nonspecific clinical manifestations and similar morphology with other organisms. A high index of suspicion is required.
Collapse
Affiliation(s)
- Aasith Villavicencio
- Division of Infectious Diseases, Department of Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Eloy F. Ruiz
- Department of Internal MedicineRutgers New Jersey Medical SchoolNewarkNew JerseyUSA
| | - Osiris Fernandez
- School of MedicinePontificia Universidad Catolica Madre y MaestraSantiago de Los CaballerosDominican Republic
| | - Anmary Fernandez
- Transplant Infectious Diseases, Infectious Disease Associates of Tampa BayTampaFloridaUSA
- Division of Infectious DiseasesUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
| | - Jose A. Gonzales‐Zamora
- Division of Infectious Diseases, Department of MedicineUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| |
Collapse
|
17
|
Brenes LR, Johnson AD, Lohse MB. Farnesol and phosphorylation of the transcriptional regulator Efg1 affect Candida albicans white-opaque switching rates. PLoS One 2023; 18:e0280233. [PMID: 36662710 PMCID: PMC9858334 DOI: 10.1371/journal.pone.0280233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 12/24/2022] [Indexed: 01/21/2023] Open
Abstract
Candida albicans is a normal member of the human microbiome and an opportunistic fungal pathogen. This species undergoes several morphological transitions, and here we consider white-opaque switching. In this switching program, C. albicans reversibly alternates between two cell types, named "white" and "opaque," each of which is normally stable across thousands of cell divisions. Although switching under most conditions is stochastic and rare, certain environmental signals or genetic manipulations can dramatically increase the rate of switching. Here, we report the identification of two new inputs which affect white-to-opaque switching rates. The first, exposure to sub-micromolar concentrations of (E,E)-farnesol, reduces white-to-opaque switching by ten-fold or more. The second input, an inferred PKA phosphorylation of residue T208 on the transcriptional regulator Efg1, increases white-to-opaque switching ten-fold. Combining these and other environmental inputs results in a variety of different switching rates, indicating that a given rate represents the integration of multiple inputs.
Collapse
Affiliation(s)
- Lucas R. Brenes
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California, United States of America
| | - Alexander D. Johnson
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California, United States of America
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, California, United States of America
| | - Matthew B. Lohse
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California, United States of America
| |
Collapse
|
18
|
Guner Ozenen G, Sahbudak Bal Z, Avcu G, Ozkaya Yazici P, Karakoyun M, Metin DY, Hilmioglu Polat S. Evaluation of candidemia in children at a university hospital: A retrospective cohort. Mycoses 2023; 66:367-377. [PMID: 36597951 DOI: 10.1111/myc.13564] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/01/2022] [Accepted: 01/02/2023] [Indexed: 01/05/2023]
Abstract
BACKGROUND Candidemia is a life-threatening infection in hospitalied children. This study aimed to evaluate candidemia's demographic and clinical characteristics and identify the risk factors and outcomes of Candida albicans (CA) and non-albicans Candida (NAC) spp. METHODS A retrospective cohort was designed to evaluate paediatric patients with candidemia between January 2008 and December 2020. RESULTS A total of 342 episodes in 311 patients were evaluated. The median age of the patients was 2.1 years (1 month-17 years and 6 months), and 59.6% were male. The prevalence of NAC (67.5%) candidemia was higher than that of CA (32.5%). The most commonly isolated Candida species was Candida parapsilosis (43.3%), followed by C. albicans (32.5%), Candida glabrata (6.1%) and Candida tropicalis (5.0%). The length of hospital stay prior to the positive culture and the total length of hospital stay were longer in the NAC group (p = .003 and p = .006). The neutrophil count was lower in the NAC group (p = .007). In the multivariate analysis, total parenteral nutrition, antifungal prophylaxis and a history of coagulase-negative staphylococci (CoNS) culture positivity in the past month were risk factors for developing candidemia due to NAC (p values were .003, .003 and .045). C. albicans and C. parapsilosis fluconazole resistance were 9.5% and 46.6%, respectively. The rates of amphotericin B resistance were 1.1% and 7.6% in C. albicans and C. parapsilosis, respectively. Mortality (14-day and 30-day) rates did not differ between the groups. CONCLUSIONS A history of CoNS culture positivity in the past month, total parenteral nutrition, and antifungal prophylaxis increases the risk of NAC candidemia.
Collapse
Affiliation(s)
- Gizem Guner Ozenen
- Division of Infectious Disease, Department of Pediatrics, Medical School of Ege University, Izmir, Turkey
| | - Zumrut Sahbudak Bal
- Division of Infectious Disease, Department of Pediatrics, Medical School of Ege University, Izmir, Turkey
| | - Gulhadiye Avcu
- Division of Infectious Disease, Department of Pediatrics, Medical School of Ege University, Izmir, Turkey
| | - Pinar Ozkaya Yazici
- Division of Intensive Care Unit, Department of Pediatrics, Medical School of Ege University, Izmir, Turkey
| | - Miray Karakoyun
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Medical School of Ege University, Izmir, Turkey
| | - Dilek Yesim Metin
- Department of Medical Microbiology, Medical School of Ege University, Izmir, Turkey
| | | |
Collapse
|
19
|
Powell LC, Adams JYM, Quoraishi S, Py C, Oger A, Gazze SA, Francis LW, von Ruhland C, Owens D, Rye PD, Hill KE, Pritchard MF, Thomas DW. Alginate oligosaccharides enhance the antifungal activity of nystatin against candidal biofilms. Front Cell Infect Microbiol 2023; 13:1122340. [PMID: 36798083 PMCID: PMC9927220 DOI: 10.3389/fcimb.2023.1122340] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/11/2023] [Indexed: 02/04/2023] Open
Abstract
Background The increasing prevalence of invasive fungal infections in immuno-compromised patients is a considerable cause of morbidity and mortality. With the rapid emergence of antifungal resistance and an inadequate pipeline of new therapies, novel treatment strategies are now urgently required. Methods The antifungal activity of the alginate oligosaccharide OligoG in conjunction with nystatin was tested against a range of Candida spp. (C. albicans, C. glabrata, C. parapsilosis, C. auris, C. tropicalis and C. dubliniensis), in both planktonic and biofilm assays, to determine its potential clinical utility to enhance the treatment of candidal infections. The effect of OligoG (0-6%) ± nystatin on Candida spp. was examined in minimum inhibitory concentration (MIC) and growth curve assays. Antifungal effects of OligoG and nystatin treatment on biofilm formation and disruption were characterized using confocal laser scanning microscopy (CLSM), scanning electron microscopy (SEM) and ATP cellular viability assays. Effects on the cell membrane were determined using permeability assays and transmission electron microscopy (TEM). Results MIC and growth curve assays demonstrated the synergistic effects of OligoG (0-6%) with nystatin, resulting in an up to 32-fold reduction in MIC, and a significant reduction in the growth of C. parapsilosis and C. auris (minimum significant difference = 0.2 and 0.12 respectively). CLSM and SEM imaging demonstrated that the combination treatment of OligoG (4%) with nystatin (1 µg/ml) resulted in significant inhibition of candidal biofilm formation on glass and clinical grade silicone surfaces (p < 0.001), with increased cell death (p < 0.0001). The ATP biofilm disruption assay demonstrated a significant reduction in cell viability with OligoG (4%) alone and the combined OligoG/nystatin (MIC value) treatment (p < 0.04) for all Candida strains tested. TEM studies revealed the combined OligoG/nystatin treatment induced structural reorganization of the Candida cell membrane, with increased permeability when compared to the untreated control (p < 0.001). Conclusions Antimicrobial synergy between OligoG and nystatin against Candida spp. highlights the potential utility of this combination therapy in the prevention and topical treatment of candidal biofilm infections, to overcome the inherent tolerance of biofilm structures to antifungal agents.
Collapse
Affiliation(s)
- Lydia C. Powell
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
- Microbiology and Infectious Disease group, Swansea University Medical School, Swansea, United Kingdom
- *Correspondence: Lydia C. Powell,
| | - Jennifer Y. M. Adams
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
| | - Sadik Quoraishi
- Otolaryngology Department, New Cross Hospital, Wolverhampton, United Kingdom
| | - Charlène Py
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
- School of Engineering, University of Angers, Angers, France
| | - Anaϊs Oger
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
- School of Engineering, University of Angers, Angers, France
| | - Salvatore A. Gazze
- Centre for Nanohealth, Swansea University Medical School, Swansea, United Kingdom
| | - Lewis W. Francis
- Centre for Nanohealth, Swansea University Medical School, Swansea, United Kingdom
| | - Christopher von Ruhland
- Central Biotechnology Services, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - David Owens
- Head and Neck Directorate, University Hospital of Wales, Cardiff, United Kingdom
| | | | - Katja E. Hill
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
| | - Manon F. Pritchard
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
| | - David W. Thomas
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
| |
Collapse
|
20
|
Wei Y, Wang Z, Liu Y, Liao B, Zong Y, Shi Y, Liao M, Wang J, Zhou X, Cheng L, Ren B. Extracellular vesicles of Candida albicans regulate its own growth through the l-arginine/nitric oxide pathway. Appl Microbiol Biotechnol 2022; 107:355-367. [PMCID: PMC9703431 DOI: 10.1007/s00253-022-12300-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/12/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Yu Wei
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Zheng Wang
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Yaqi Liu
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Binyou Liao
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Yawen Zong
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Yangyang Shi
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Min Liao
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Jiannan Wang
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| | - Biao Ren
- State Key Laboratory of Oral Diseases &, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610000 Sichuan Province China
| |
Collapse
|
21
|
Jahanshiri Z, Manifar S, Arastehnazar F, Hatami F, Lotfali E. Azole Resistance in Candida albicans Isolates from Oropharyngeal Candidiasis is Associated with ERG11 Mutation and Efflux Overexpression. Jundishapur J Microbiol 2022; 15. [DOI: 10.5812/jjm-131046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/08/2022] [Accepted: 10/25/2022] [Indexed: 01/03/2025] Open
Abstract
Background: Azole resistance rates are rising in Candida species. Fluconazole is one of the most important antifungal drugs used in candidiasis treatment. Objectives: We identified the molecular mechanisms of fluconazole resistance of Candida albicans oropharyngeal candidiasis (OPC) isolates obtained from head and neck cancer patients, a study carried out between 2018 and 2020. Methods: One hundred and twenty-five C. albicans clinical isolates were collected. Antifungal susceptibilities were determined by the CLSI- M27-A3 method. The ERG11 gene was amplified and sequenced to discover SNP mutation. Moreover, real-time PCR was carried out to measure the mRNA levels of ERG11, CDR1, CDR2, and MDR1. Results: Resistance to fluconazole was found in 15 C. albicans isolates. Amino acid substitutions E266D and D116E were observed in resistant, sensitive dose-dependent (SDD), and susceptible C. albicans isolates. K128T, G465S, A114S, Y257H and V488I were in relation to fluconazole resistance. D504A, P375A, W520C, G59S, and V51L were novel substitutions detected in the isolates; except for D504A, other mutations were observed only in resistance isolates. The expression levels of CDR2, CDR1, MDR1, and ERG11 were increased compared to susceptible isolates, respectively. Conclusions: ERG11 mutation was the principal mechanism for fluconazole resistance in C. albicans isolated from oropharyngeal candidiasis patients, and caspofungin can be used as the effective antifungal substance in fluconazole resistance situation for C. albicans infection.
Collapse
|
22
|
Wang Y, Zhou H, Wang W, Duan N, Luo Z, Chai H, Jiang L, Chen Q, Liu J, Hua H, Yan Z, Fan Y, Xu J, Guan X, Wang H, Lu H, Lun W, Fei W, Zhang T, Zhao J, Jia C, Kong H, Shen X, Liu Q, Wang W, Tang G. Efficacy and safety of miconazole muco-adhesive tablet versus itraconazole in oropharyngeal candidiasis: A randomized, multi-centered, double-blind, phase 3 trial. Med Mycol 2022; 60:myac076. [PMID: 36149321 DOI: 10.1093/mmy/myac076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 06/16/2022] [Accepted: 09/21/2022] [Indexed: 11/14/2022] Open
Abstract
Oropharyngeal candidiasis (OPC) is an opportunistic infection treated with anti-fungal agents. Herein, we evaluate the efficacy and safety of miconazole buccal tablets (MBT) and itraconazole capsules in the localized treatment of patients with OPC. In this multi-centered, double-blinded, phase III trial (CTR20130414), both males and non-pregnant females (≥18 years) with OPC were randomized (1:1) to MBT plus placebo (experimental group) or itraconazole capsules plus placebo (control group). The primary endpoint was clinical cure at the end-of-treatment period [visit 4 (V4)] while secondary endpoints were clinical remission rates, partial remission rates, mycological cure, clinical relapse, and adverse events (AEs). All endpoints were statistically analyzed in both the full analysis set (FAS) and per-protocol (PP) set. A total of 431 (experimental: 216; control: 215) subjects were included. At V4, in the FAS set, the clinical cure was achieved in 68% and 59% patients in experimental and control groups, respectively with a treatment difference of 9% [95% confidence interval (CI): -1,19; P < .001] demonstrating non-inferiority of MBT over itraconazole. At V4, mycological cure rates were 68.2% and 42.0% in the experimental group and control groups (P < .001), respectively in FAS. The relapse rates were 5.4% and 6.6%, respectively, in the experimental and control groups. A total of 210 patients experienced AEs during treatment with 47.7% in the experimental group and 49.8% in the control group with no deaths. This study demonstrated that once-daily treatment with MBT was non-inferior to itraconazole with higher mycological cure rates and was tolerable with mild AE in patients with OPC.
Collapse
Affiliation(s)
- Yufeng Wang
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Stomatological Center; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory, Shanghai 200011, China
| | - Haiwen Zhou
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Stomatological Center; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory, Shanghai 200011, China
| | - Wenmei Wang
- Department of Oral Medicine, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, Jiangsu, China
| | - Ning Duan
- Department of Oral Medicine, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, Jiangsu, China
| | - Zhixiao Luo
- Dental Department, Taihe Hospital, Affiliated Hospital of Hubei, University of Medicine, Shiyan 442099, Hubei, China
| | - Hongbo Chai
- Dental Department, Taihe Hospital, Affiliated Hospital of Hubei, University of Medicine, Shiyan 442099, Hubei, China
| | - Lu Jiang
- Department of Oral Medicine, West China School/Hospital Stomatology Sichuan University, Chengdu 610042, Sichuan, China
| | - Qianming Chen
- Department of Oral Medicine, West China School/Hospital Stomatology Sichuan University, Chengdu 610042, Sichuan, China
| | - Jinli Liu
- Department of Oral Medicine, West China School/Hospital Stomatology Sichuan University, Chengdu 610042, Sichuan, China
| | - Hong Hua
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Zhimin Yan
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Yuan Fan
- Department of Oral Medicine, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Juanyong Xu
- Department of Oral Medicine, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Xiaobing Guan
- Department of Oral Medicine, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Hongjian Wang
- Department of Oral Medicine, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Hongzhou Lu
- Department of Infectious Diseases and Immunology, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Wenhui Lun
- Department of Dermatology and Venereology, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Wei Fei
- Department of Stomatology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu 610072, Sichuan, China
| | - Tong Zhang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Jizhi Zhao
- Department of Stomatology, Peking Union Medical College Hospital, Beijing 100730, China
| | - Chunling Jia
- Department of Oral Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Hui Kong
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Oral Medicine, Department of Operative Dentistry & Endodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Xuemin Shen
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Stomatological Center; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory, Shanghai 200011, China
| | - Qing Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Oral Medicine, Department of Operative Dentistry & Endodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Weizhi Wang
- Department of Oral Medicine, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Guoyao Tang
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Stomatological Center; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory, Shanghai 200011, China
| |
Collapse
|
23
|
Abstract
Infectious esophagitis is the third most common cause of esophagitis after gastroesophageal reflux disease and eosinophilic esophagitis (EoE) and should always be considered in the differential of patients with dysphagia and odynophagia. The most common organisms causing disease are candida, Herpes simplex virus (HSV) and cytomegalovirus (CMV). It is well recognized that an impaired local or systemic immune system is a risk factor for disease; however, esophageal dysmotility and disruptions in esophageal homeostasis and the esophageal milieu are likely to represent additional risk factors in disease pathogenesis.
Collapse
|
24
|
Singh M, Verma H, Bhandu P, Kumar M, Narendra G, Choudhary S, Singh PK, Silakari O. Network Analysis Guided Designing of Multi-Targeted Anti-Fungal Agents: Synthesis and Biological Evaluation. J Mol Struct 2022; 1272:134128. [PMID: 36101882 PMCID: PMC9458262 DOI: 10.1016/j.molstruc.2022.134128] [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: 04/13/2022] [Revised: 08/20/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022]
Abstract
During the ongoing pandemic, there have been increasing reports of invasive fungal disease (IFD), particularly among immunocompromised populations. Candida albicans is one of the most common clinical pathogenic microorganisms which have become a serious health threat to population either infected with Covid-19 or on treatment with immunosuppressant's/broad-range antibiotics. Currently, benzothiazole is a well explored scaffold for anti-fungal activity, especially mercapto substituted benzothiazoles. It is reported that exploring the 2nd position of benzothiazoles yield improved anti-fungal molecules. Therefore, in the current study, lead optimization approach using bioisosteric replacement protocol was followed to improve the anti-fungal activity of an already reported benzothiazole derivative, N-(1,3-benzothiazole-2-yl)-2-(pyridine-3-ylformohydrazido) acetamide. To rationally identify the putative anti-candida targets of this derivative, network analysis was carried out. Complexes of designed compounds and identified putative targets were further analyzed for the docking interactions and their consequent retention after the completion of exhaustive MD simulations. Top seven designed compounds were synthesized and evaluated for in-vitro anti-fungal property against Candida, which indicated that compounds 1.2c and 1.2f possess improved and comparable anti-fungal activity to N-(1,3-benzothiazole-2-yl)-2-(pyridine-3-ylformohydrazido) acetamide and Nystatin, respectively.
Collapse
Affiliation(s)
- Manmeet Singh
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Himanshu Verma
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Priyanka Bhandu
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Manoj Kumar
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Gera Narendra
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Shalki Choudhary
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Pankaj Kumar Singh
- Faculty of Medicine, Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, FI-20014, Finland
| | - Om Silakari
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| |
Collapse
|
25
|
Holzknecht J, Dubrac S, Hedtrich S, Galgóczy L, Marx F. Small, Cationic Antifungal Proteins from Filamentous Fungi Inhibit Candida albicans Growth in 3D Skin Infection Models. Microbiol Spectr 2022; 10:e0029922. [PMID: 35499318 PMCID: PMC9241769 DOI: 10.1128/spectrum.00299-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/01/2022] [Indexed: 12/19/2022] Open
Abstract
The emerging resistance of human-pathogenic fungi to antifungal drugs urges the development of alternative therapeutic strategies. The small, cationic antifungal proteins (AFPs) from filamentous ascomycetes represent promising candidates for next-generation antifungals. These bio-molecules need to be tested for tolerance in the host and efficacy against fungal pathogens before they can be safely applied in humans. Testing of the efficacy and possible adverse effects of new drug candidates in three-dimensional (3D) human-cell based models represents an advantageous alternative to animal experiments. In, this study, as a proof-of-principle, we demonstrate the usefulness of 3D skin infection models for screening new antifungal drug candidates for topical application. We established a cutaneous infection with the opportunistic human-pathogenic yeast Candida albicans in a commercially available 3D full-thickness (FT) skin model to test the curative potential of distinct AFPs from Penicillium chrysogenum (PAFopt, PAFB, and PAFC) and Neosartorya (Aspergillus) fischeri (NFAP2) in vitro. All tested AFPs were comparably well tolerated by the skin models. The infected 3D models exhibited reduced epidermal permeability barriers, allowing C. albicans to colonize the epidermal and dermal layers, and showed increased secretion of the pro-inflammatory cytokine IL-6 and the chemokine IL-8. AFP treatment diminished the fungal burden and penetration depth of C. albicans in the infected models. The epidermal permeability barrier was restored and the secretion of IL-8 was decreased following AFP treatment. In summary, our study proves that the tested AFPs exhibit antifungal potential against cutaneous C. albicans infection in a 3D FT skin model. IMPORTANCE Candida albicans represents one of the most prevalent opportunistic fungal pathogens, causing superficial skin and mucosal infections in humans with certain predisposing health conditions and life-threatening systemic infections in immunosuppressed patients. The emerging drug resistance of this human-pathogenic yeast and the limited number of antifungal drugs for prevention and treatment of infections urgently demands the identification of new antifungal compounds with novel mechanisms of action. Small, cationic antifungal proteins (AFPs) from filamentous fungi represent promising candidates for next-generation antifungals for topical application. These bio-molecules need to be tested for tolerance by the host and efficacy in pathogen clearance prior to being involved in clinical trials. In a proof-of-principle study, we provide evidence for the suitability of 3D human-cell based models as advantageous alternatives to animal experiments. We document the tolerance of specific AFPs and their curative efficacy against cutaneous C. albicans infection in a 3D skin model.
Collapse
Affiliation(s)
- Jeanett Holzknecht
- Biocenter, Institute of Molecular Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sandrine Dubrac
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sarah Hedtrich
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - László Galgóczy
- Department of Biotechnology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
- Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
| | - Florentine Marx
- Biocenter, Institute of Molecular Biology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
26
|
Hernandez-Cuellar E, Guerrero-Barrera AL, Avelar-Gonzalez FJ, Díaz JM, Santiago ASD, Chávez-Reyes J, Poblano-Sánchez E. Characterization of Candida albicans and Staphylococcus aureus polymicrobial biofilm on different surfaces. Rev Iberoam Micol 2022; 39:36-43. [PMID: 35738989 DOI: 10.1016/j.riam.2022.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 03/24/2022] [Accepted: 04/21/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Staphylococcus aureus and Candida albicans have been co-isolated from biofilm-associated diseases such as denture stomatitis, periodontitis, and burn wound infections, as well as from medical devices. However, the polymicrobial biofilm of both microorganisms has not been fully characterized. AIMS To characterize the polymicrobial biofilm of C. albicans and S. aureus in terms of microbial density, synergy, composition, structure, and stability against antimicrobials and chemical agents. METHODS Crystal violet assay was used to measure the biofilm formation. Scanning electron microscopy and confocal microscopy were used to analyze the structure and chemical composition of the biofilms, respectively. RESULTS Supplemented media with fetal bovine serum (FBS) decreased the biofilm formation of S. aureus and the polymicrobial biofilm. For C. albicans, depending on the culture media, the addition of glucose or FBS had a positive effect in biofilm formation. FBS decreased the adhesion to polystyrene wells for both microorganisms. Supplementing the media with glucose and FBS enhanced the growth of C. albicans and S. aureus, respectively. It seems that C. albicans contributes the most to the adhesion process and to the general structure of the biofilms on all the surfaces tested, including a catheter model. Interestingly, S. aureus showed a great adhesion capacity to the surface of C. albicans in the biofilms. Proteins and β-1,6-linked polysaccharides seem to be the most important molecules in the polymicrobial biofilm. CONCLUSIONS The polymicrobial biofilm had a complex structure, with C. albicans serving as a scaffold where S. aureus adheres, preferentially to the hyphal form of the fungus. Detection of polymicrobial infections and characterization of biofilms will be necessary in the future to provide a better treatment.
Collapse
Affiliation(s)
- Eduardo Hernandez-Cuellar
- Laboratorio de Biología Celular y Tisular, Departamento de Morfología, Universidad Autónoma de Aguascalientes (UAA), Aguascalientes, Mexico.
| | - Alma Lilián Guerrero-Barrera
- Laboratorio de Biología Celular y Tisular, Departamento de Morfología, Universidad Autónoma de Aguascalientes (UAA), Aguascalientes, Mexico
| | - Francisco Javier Avelar-Gonzalez
- Laboratorio de Ciencias Ambientales, Departamento de Fisiología y Farmacología, Universidad Autónoma de Aguascalientes (UAA), Aguascalientes, Mexico
| | - Juan Manuel Díaz
- Laboratorio de Biología Celular y Tisular, Departamento de Morfología, Universidad Autónoma de Aguascalientes (UAA), Aguascalientes, Mexico
| | - Alfredo Salazar de Santiago
- Unidad Académica de Odontología, Área de Ciencias de la Salud, Universidad Autónoma de Zacatecas, Zacatecas, Mexico
| | - Jesús Chávez-Reyes
- Laboratorio de Farmacología y Terapéutica Experimental, Departamento de Fisiología y Farmacología, Universidad Autónoma de Aguascalientes (UAA), Aguascalientes, Mexico
| | - Emanuel Poblano-Sánchez
- Institute for Social Security and Services for State Workers (ISSSTE), Aguascalientes, Mexico
| |
Collapse
|
27
|
Boya BR, Lee JH, Lee J. Antibiofilm and Antimicrobial Activities of Chloroindoles Against Uropathogenic Escherichia coli. Front Microbiol 2022; 13:872943. [PMID: 35783430 PMCID: PMC9244173 DOI: 10.3389/fmicb.2022.872943] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/23/2022] [Indexed: 12/30/2022] Open
Abstract
Uropathogenic Escherichia coli (UPEC) is a nosocomial pathogen associated with urinary tract infections and expresses several virulence factors that cause recurring infections and cystitis of the bladder, which can lead to pyelonephritis. UPEC uses different types of extracellular appendages like fimbriae and pili that aid colonization and adherence to bladder epithelium and can form persistent biofilm-like bacterial communities that aid its survival after the deployment of host immune responses. We investigated the antibiofilm, antimicrobial, and antivirulence properties of three indole derivatives namely, 4-chloroindole, 5-chloroindole, and 5-chloro 2-methyl indole. All the three chloroindoles had MICs of 75 μg/ml and inhibited biofilm formation by an average of 67% at 20 μg/ml. In addition, they inhibited swarming and swimming motilities, which are essential for dissemination from bacterial communities and colonization, reduced cell surface hydrophobicity, and inhibited indole production and curli formation. Gene expression analysis showed all three chloroindoles significantly downregulated the expressions of virulence genes associated with adhesion, stress regulation, and toxin production. A 3D-QSAR analysis revealed substitutions at the fourth and fifth positions of the indole moiety favored antimicrobial activity. Furthermore, these chloroindoles potently inhibited biofilm formation in other nosocomial pathogens and polymicrobial consortia.
Collapse
|
28
|
Buakaew W, Pankla Sranujit R, Noysang C, Krobthong S, Yingchutrakul Y, Thongsri Y, Potup P, Daowtak K, Usuwanthim K. Proteomic Analysis Reveals Proteins Involved in the Mode of Action of β-Citronellol Identified From Citrus hystrix DC. Leaf Against Candida albicans. Front Microbiol 2022; 13:894637. [PMID: 35677908 PMCID: PMC9168680 DOI: 10.3389/fmicb.2022.894637] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 04/26/2022] [Indexed: 11/23/2022] Open
Abstract
Candida albicans is a fungus that lives primarily on the mucosal surfaces of healthy humans, such as the oral cavity, vagina, and gastrointestinal tract. This commensal organism can be controlled by other microbiota, while certain conditions can increase the risk of C. albicans outgrowth and cause disease. Prevalence of the drug-resistant phenotype, as well as the severity of C. albicans infection in immunocompromised patients, presents a challenge for scientists to develop novel, effective treatment, and prevention strategies. β-Citronellol is an intriguing active compound of several plants that has been linked to antifungal activity, but data on the mechanism of action in terms of proteomic profiling are lacking. Here, β-citronellol identified from Citrus hystrix DC. leaf against C. albicans were evaluated. A proteomic approach was used to identify potential target proteins involved in the mode of action of β-citronellol. This study identified and discussed three protein groups based on the 126 major proteins that were altered in response to β-citronellol treatment, 46 of which were downregulated and 80 of which were upregulated. Significant protein groups include cell wall proteins (e.g., Als2p, Rbt1p, and Pga4p), cellular stress response enzymes (e.g., Sod1p, Gst2p, and Ddr48p), and ATP synthesis-associated proteins (e.g., Atp3p, Atp7p, Cox1p, and Cobp). Results demonstrated the complexities of protein interactions influenced by β-citronellol treatment and highlighted the potential of antifungal activity for future clinical and drug development research.
Collapse
Affiliation(s)
- Watunyoo Buakaew
- Cellular and Molecular Immunology Research Unit, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| | - Rungnapa Pankla Sranujit
- Faculty of Integrative Medicine, Rajamangala University of Technology Thanyaburi, Pathum Thani, Thailand
| | - Chanai Noysang
- Faculty of Integrative Medicine, Rajamangala University of Technology Thanyaburi, Pathum Thani, Thailand
| | - Sucheewin Krobthong
- Interdisciplinary Graduate Program in Genetic Engineering, Kasetsart University, Bangkok, Thailand
- Center for Neuroscience, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Yodying Yingchutrakul
- Center for Neuroscience, Faculty of Science, Mahidol University, Bangkok, Thailand
- National Omics Center, National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Yordhathai Thongsri
- Cellular and Molecular Immunology Research Unit, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| | - Pachuen Potup
- Cellular and Molecular Immunology Research Unit, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| | - Krai Daowtak
- Cellular and Molecular Immunology Research Unit, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| | - Kanchana Usuwanthim
- Cellular and Molecular Immunology Research Unit, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| |
Collapse
|
29
|
Early Empirical Anidulafungin Reduces the Prevalence of Invasive Candidiasis in Critically Ill Patients: A Case-control Study. J Crit Care Med (Targu Mures) 2022; 8:89-99. [PMID: 35950155 PMCID: PMC9097641 DOI: 10.2478/jccm-2022-0006] [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: 01/13/2022] [Accepted: 04/06/2022] [Indexed: 11/21/2022] Open
Abstract
Introduction Invasive candidiasis (IC) in critically ill patients is a serious infection with high rate of mortality. As an empirical therapy, like antibiotics, the use of antifungals is not common in intensive care units (ICUs) worldwide. The empirical use of echinocandins including anidulafungin is a recent trend. Aim of the study The objective of this study was to assess the impact of empirical anidulafungin in the development of invasive candidiasis in critically ill patients in ICU. Methods This retrospective case-control study was conducted on 149 patients with sepsis with/without septic shock and bacterial pneumonia. All the patients were divided into two groups. The ‘control group’ termed as ‘NEAT group’ received no empirical anidulafungin therapy and the ‘treated group’ termed as ‘EAT group’ received empirical anidulafungin therapy in early hospitalization hours. Results Seventy-two and 77 patients were divided into the control and the treated group, respectively. Patients in EAT group showed less incidences of IC (5.19%) than that of the NEAT group (29.17%) (p = 0.001). Here, the relative risk (RR) was 0.175 (95% CI, 0.064-0.493) and the risk difference (RD) rate was 24% (95% CI, 12.36%-35.58%). The 30-day all-cause mortality rate in NEAT group was higher (19.44%) than that of in EAT group (10.39%) (p = 0.04). Within the first 10-ICU-day, patients in the EAT group left ICU in higher rate (62.34%) than that in the NEAT group (54.17%). Conclusion Early empirical anidulafungin within 6 h of ICU admission reduced the risk of invasive candidiasis, 30-day all-cause mortality rate and increased ICU leaving rate within 10-day of ICU admission in critically ill patients.
Collapse
|
30
|
Castanheira M, Deshpande LM, Davis AP, Carvalhaes CG, Pfaller MA. Azole Resistance in Candida glabrata Clinical Isolates from Global Surveillance is Associated with Efflux Overexpression. J Glob Antimicrob Resist 2022; 29:371-377. [DOI: 10.1016/j.jgar.2022.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 11/28/2022] Open
|
31
|
Dong P, Zhan Y, Jusuf S, Hui J, Dagher Z, Mansour MK, Cheng J. Photoinactivation of Catalase Sensitizes Candida albicans and Candida auris to ROS-Producing Agents and Immune Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104384. [PMID: 35119220 PMCID: PMC8981478 DOI: 10.1002/advs.202104384] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 01/07/2022] [Indexed: 06/14/2023]
Abstract
Microbes have developed their own specific strategies to cope with reactive oxygen species (ROS). Catalase, a heme-containing tetramer expressed in a broad range of aerobic fungi, shows remarkable efficiency in degrading hydrogen peroxide (H2 O2 ) for fungal survival and host invasion. Here, it is demonstrated that catalase inactivation by blue light renders fungal cells highly susceptible to ROS attack. To confirm catalase as a major molecular target of blue light, wild type Candida albicans are systematically compared with a catalase-deficient mutant strain regarding their susceptibility to ROS through 410 nm treatment. Upon testing a wide range of fungal species, it is found that intracellular catalase can be effectively and universally inactivated by 410 nm blue light. It is also found that photoinactivation of catalase in combination with ROS-generating agents is highly effective in total eradication of various fungal species, including multiple Candida auris strains, the causative agent of the global fungal epidemic. In addition, photoinactivation of catalase is shown to facilitate macrophage killing of intracellular Candida albicans. The antifungal efficacy of catalase photoinactivation is further validated using a C. albicans-induced mouse model of skin abrasion. Taken together, the findings offer a novel catalase-photoinactivation approach to address multidrug-resistant Candida infections.
Collapse
Affiliation(s)
- Pu‐Ting Dong
- Department of Biomedical EngineeringBoston UniversityBostonMA02215USA
- Photonics CenterBoston UniversityBostonMA02215USA
| | - Yuewei Zhan
- Department of Biomedical EngineeringBoston UniversityBostonMA02215USA
- Photonics CenterBoston UniversityBostonMA02215USA
| | - Sebastian Jusuf
- Department of Biomedical EngineeringBoston UniversityBostonMA02215USA
- Photonics CenterBoston UniversityBostonMA02215USA
| | - Jie Hui
- Department of Biomedical EngineeringBoston UniversityBostonMA02215USA
- Photonics CenterBoston UniversityBostonMA02215USA
| | - Zeina Dagher
- Division of Infectious DiseasesMassachusetts General HospitalBostonMA02114USA
- Harvard Medical SchoolBostonMA02115USA
| | - Michael K. Mansour
- Division of Infectious DiseasesMassachusetts General HospitalBostonMA02114USA
- Harvard Medical SchoolBostonMA02115USA
| | - Ji‐Xin Cheng
- Department of Biomedical EngineeringBoston UniversityBostonMA02215USA
- Photonics CenterBoston UniversityBostonMA02215USA
| |
Collapse
|
32
|
Mashaly GES, Zeid MS. Candida albicans Genotyping and Relationship of Virulence Factors with Fluconazole Tolerance in Infected Pediatric Patients. Infect Drug Resist 2022; 15:2035-2043. [PMID: 35480061 PMCID: PMC9037425 DOI: 10.2147/idr.s344998] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 02/05/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Ghada El-Saeed Mashaly
- Medical Microbiology and Immunology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Correspondence: Ghada El-Saeed Mashaly, Faculty of Medicine, Mansoura University, Box 50, Al- Mansoura, 35516, Egypt, Tel +201003062542, Email
| | - Mayada Sabry Zeid
- Infectious Diseases and Malnutrition, Pediatrics Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
33
|
Hillenbrand M, Mendy A, Patel K, Wilkinson R, Liao S, Robertson J, Apewokin S. The Incidence of Ocular Complications in Candidemic Patients and Implications for the Practice of Routine Eye Exams. Open Forum Infect Dis 2022; 9:ofac045. [PMID: 35355893 PMCID: PMC8962754 DOI: 10.1093/ofid/ofac045] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/25/2022] [Indexed: 11/26/2022] Open
Abstract
Background Ocular candidiasis is a known complication of candidemia. Given the poor ocular penetration of echinocandins, there is some concern that the increasing use of echinocandins may portend an increased incidence of ophthalmic complications. We examined the changing trends in antifungal prescribing patterns and the incidence of ophthalmic complications after candidemia. Methods Patients with blood cultures positive for Candida species between January 2014 and June 2020 who underwent screening fundoscopic examination by an ophthalmologist were analyzed. The χ2 analysis was used to compare antifungal prescriptions and ocular exam findings before and after 2016. Trend analysis was also performed to assess temporal changes in prescribing practices and eye exam findings. Results There were 226 candidemia cases during the study period, 129 (57.1%) of which underwent screening eye exams. From 2014 to 2015, 24 of 37 (64.5%) patients received eye-penetrating antifungals compared to 36 of 92 (39.1%) from 2016 to 2020 (P = .008). Overall, 30 of 129 (23.3%) patients had abnormal eye exams with the prevalence of abnormal findings being 7 of 37 (18.9%) before 2016 compared to 23 of 92 (25%, P = .46) thereafter. A trend analysis revealed an increase in abnormal eye findings over the study period (P = .008). Of the 30 patients who had abnormal eye exams, 9 (30%) had a change in systemic antifungal therapy from echinocandins to eye-penetrating antifungals. Echinocandin use was associated with abnormal eye findings. Conclusions Prescription of eye-penetrating antifungals for candidemia has trended down since 2016. This was associated with a concomitant increase in abnormal findings on screening fundoscopy. Abnormal eye exams were not uncommon throughout our study period.
Collapse
Affiliation(s)
- Molly Hillenbrand
- Department of Internal Medicine, Duke University , Durham, North Carolina, USA
| | - Angelico Mendy
- Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Kavya Patel
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Racheal Wilkinson
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Siyun Liao
- University of Cincinnati College of Pharmacy, Cincinnati, Ohio , USA
| | - Jamie Robertson
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Senu Apewokin
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
34
|
Katta T, Tavakoli K. Necrotizing Cholecystitis in the Gallbladder: A Case Report. Cureus 2022; 14:e21368. [PMID: 35070585 PMCID: PMC8764969 DOI: 10.7759/cureus.21368] [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] [Accepted: 01/04/2022] [Indexed: 11/13/2022] Open
Abstract
Infections caused by Candida species have shown a considerable increase in frequency in the recent past, and hence they are a cause of significant concern among medical practitioners. There are many factors that contribute to the occurrence of Candida-related infections in particular groups of patients. In this report, we present a case that highlights the causes and appropriate treatment methods of the condition. Patients with acute necrotizing cholecystitis often show poor outcomes after treatment, and hence physicians need to be alert when dealing with patients with this condition and should provide the best treatment method. We report a case of necrotizing cholecystitis in a 55-year-old female with a medical history of cholelithiasis, obesity, seizures, cocaine abuse, and anemia. She also reported lower abdominal pain, felt bloated, and complained of headache, dizziness, lack of appetite, shortness of breath, and vomiting. The patient underwent several lab tests as well as a CT scan of the abdomen, hepatobiliary iminodiacetic acid (HIDA) scan, endoscopy, and cholecystectomy.
Collapse
|
35
|
Herman A, Herman AP. Could Candida Overgrowth Be Involved in the Pathophysiology of Autism? J Clin Med 2022; 11:442. [PMID: 35054136 PMCID: PMC8778531 DOI: 10.3390/jcm11020442] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/31/2021] [Accepted: 01/13/2022] [Indexed: 02/05/2023] Open
Abstract
The purpose of this review is to summarize the current acquiredknowledge of Candida overgrowth in the intestine as a possible etiology of autism spectrum disorder (ASD). The influence of Candida sp. on the immune system, brain, and behavior of children with ASD isdescribed. The benefits of interventions such as a carbohydrates-exclusion diet, probiotic supplementation, antifungal agents, fecal microbiota transplantation (FMT), and microbiota transfer therapy (MTT) will be also discussed. Our literature query showed that the results of most studies do not fully support the hypothesis that Candida overgrowth is correlated with gastrointestinal (GI) problems and contributes to autism behavioral symptoms occurrence. On the one hand, it was reported that the modulation of microbiota composition in the gut may decrease Candida overgrowth, help reduce GI problems and autism symptoms. On the other hand, studies on humans suggesting the beneficial effects of a sugar-free diet, probiotic supplementation, FMT and MTT treatment in ASD are limited and inconclusive. Due to the increasing prevalence of ASD, studies on the etiology of this disorder are extremely needed and valuable. However, to elucidate the possible involvement of Candida in the pathophysiology of ASD, more reliable and well-designed research is certainly required.
Collapse
Affiliation(s)
- Anna Herman
- Faculty of Health Sciences, Warsaw School of Engineering and Health, Bitwy Warszawskiej 20 18, 19 Street, 02-366 Warsaw, Poland
| | - Andrzej Przemysław Herman
- Department of Genetic Engineering, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3 Street, 05-110 Jabłonna, Poland;
| |
Collapse
|
36
|
Chotiprasitsakul D, Kijnithikul A, Uamkhayan A, Santanirand P. Predictive Value of Urinalysis and Recent Antibiotic Exposure to Distinguish Between Bacteriuria, Candiduria, and No-Growth Urine. Infect Drug Resist 2021; 14:5699-5709. [PMID: 35002261 PMCID: PMC8722576 DOI: 10.2147/idr.s343021] [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: 10/06/2021] [Accepted: 12/08/2021] [Indexed: 11/25/2022] Open
Abstract
PURPOSE Urinary tract infections are diagnosed by clinical symptoms and detection of causative uropathogen. Antibiotics are usually not indicated in candiduria and no-growth urine. We aimed to develop a predictive score to distinguish bacteriuria, candiduria, and no-growth urine, and to describe the distribution of microorganisms in urine. PATIENTS AND METHODS A single-center, retrospective cohort study was conducted between January 2017 and November 2017. Patients with concomitant urinalysis and urine culture were randomly sorted for a clinical prediction model. Multivariable regression analysis was performed to determine factors associated with bacteriuria, candiduria, and no-growth urine. A scoring system was constructed by rounding the regression coefficient for each predictor to integers. Accuracy of the score was measured by the concordance index (c-index). RESULTS There were 8091 positive urine cultures: bacteria 85.6%, Candida 13.7%. Randomly selected cases were sorted into derivation and validation cohorts (448 cases and 272 cases, respectively). Numerous yeast on urinalysis predicted candiduria with complete accuracy; therefore, it was excluded from a score construction. We developed a NABY score based on: positive nitrite, 1 point; Antibiotic exposure within 30 days, -2 points; numerous Bacteria in urine, 2 points; few Yeast in urine, -2 points; moderate Yeast in urine, -5 points. The c-index was 0.85 (derivation) and 0.82 (validation). A score ≥0 predicted 76% and 54% of bacteriuria in the derivation and validation cohorts, respectively. A score ≤-3 predicted 96% of candiduria in both cohorts. CONCLUSION Numerous yeast on urinalysis and the NABY score may help identify patients with a low risk of bacteriuria in whom empiric antibiotics for UTIs can be avoided.
Collapse
Affiliation(s)
- Darunee Chotiprasitsakul
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Akara Kijnithikul
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Anuchat Uamkhayan
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Pitak Santanirand
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
37
|
Ghosh A, Sarkar A, Paul P, Patel P. The rise in cases of mucormycosis, candidiasis and aspergillosis amidst COVID19. FUNGAL BIOL REV 2021; 38:67-91. [PMID: 34548877 PMCID: PMC8445778 DOI: 10.1016/j.fbr.2021.09.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 09/05/2021] [Accepted: 09/08/2021] [Indexed: 12/15/2022]
Abstract
The Coronavirus outbreak globally has changed the medical system and also led to a shortage of medical facilities in both developing and underdeveloped countries. The COVID19 disease, being novel in nature along with high infectivity and frequent mutational rate, has been termed to be fatal across the globe. The advent of infection by SARS-CoV-2 has brought a myriad of secondary complications and comorbidities resulting in additional challenges to the health care system induced by novel therapeutic procedures. The emerging variant with respect to the Indian subcontinent and the associated genetic mutations have worsened the situation at hand. Proper clinical management along with epidemiological studies and clinical presentations in scientific studies and trials is necessary in order to combat the simultaneous waves of emerging strains. This article summarizes three of the major fungal outbreaks in India namely mucormycosis, candidiasis and aspergillosis, and elaborates their subtypes, pathogenesis, symptoms and treatment and detection techniques. A detail of future therapeutics under consideration are also elaborated along with a general hypothesis on how COVID19 is related to immunological advances leading to major widespread fungal infection in the country. The factors that contribute in promoting virus proliferation and invasive fungal infections include cell-mediated immunity, associated immunocompromised conditions and treatment protocols that slows down immune mechanisms. To better comprehend a fungal or bacterial outbreak, it is very important to conduct audits mediated through multicenter national and state research teams for recognizing patterns and studying current cases of fungal infection in both healthy and comorbid groups of COVID19 patients.
Collapse
Affiliation(s)
- Asmita Ghosh
- Department of Biotechnology, Heritage Institute of Technology, Kolkata 700107, West Bengal, India
| | - Anusua Sarkar
- Department of Biotechnology, Heritage Institute of Technology, Kolkata 700107, West Bengal, India
| | - Pubali Paul
- Department of Biotechnology, Heritage Institute of Technology, Kolkata 700107, West Bengal, India
| | - Parth Patel
- H. K. College of Pharmacy, Jogeshwari West, Mumbai 400102, Maharashtra, India
| |
Collapse
|
38
|
Economic Evaluation of Budesonide Orodispersible Tablets for the Treatment of Eosinophilic Esophagitis: A Cost-Utility Analysis. Adv Ther 2021; 38:5737-5751. [PMID: 34699003 DOI: 10.1007/s12325-021-01957-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/08/2021] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Budesonide orodispersible tablets (BOT) have been approved in Europe and Canada for the treatment of eosinophilic esophagitis (EoE), a rare and chronic disease. The objective of this study was to assess the economic impact of BOT on both the induction and maintenance of clinico-pathological remission of EoE by performing a cost-utility analysis (CUA). METHODS For both the induction and maintenance settings, BOT was compared to no treatment in a target population of adult patients with EoE non-responsive to proton pump inhibitor (PPI) treatment. Markov models were developed for the induction and maintenance settings over 52-week and life-time horizons, respectively. Analyses were performed from both a Canadian Ministry of Health (MoH) and societal perspective. The resulting incremental cost-utility ratios (ICURs) were compared to a willingness-to-pay (WTP) threshold of $50,000 Canadian dollars/quality-adjusted life-year (QALY). Sensitivity and scenario analyses were conducted to assess the robustness of the base-case results. RESULTS In the base-case probabilistic analysis, BOT compared to no treatment resulted in an ICUR of $1073/QALY and $30,555/QALY from a MoH perspective in the induction and maintenance settings, respectively. BOT was a cost-effective option for both induction and maintenance in > 99% of Monte Carlo simulations. In the scenario analyses, the deterministic ICUR of BOT compared to no treatment varied from $682/QALY to $8510/QALY in the induction setting and $21,005/QALY to $55,157/QALY in the maintenance setting. CONCLUSION BOT was cost-effective compared to no treatment for both the induction and maintenance of clinico-pathological remission of EoE in patients non-responsive to PPIs.
Collapse
|
39
|
The antimicrobial and immunomodulatory effects of Ionophores for the treatment of human infection. J Inorg Biochem 2021; 227:111661. [PMID: 34896767 DOI: 10.1016/j.jinorgbio.2021.111661] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 10/27/2021] [Accepted: 11/03/2021] [Indexed: 12/18/2022]
Abstract
Ionophores are a diverse class of synthetic and naturally occurring ion transporter compounds which demonstrate both direct and in-direct antimicrobial properties against a broad panel of bacterial, fungal, viral and parasitic pathogens. In addition, ionophores can regulate the host-immune response during communicable and non-communicable disease states. Although the clinical use of ionophores such as Amphotericin B, Bedaquiline and Ivermectin highlight the utility of ionophores in modern medicine, for many other ionophore compounds issues surrounding toxicity, bioavailability or lack of in vivo efficacy studies have hindered clinical development. The antimicrobial and immunomodulating properties of a range of compounds with characteristics of ionophores remain largely unexplored. As such, ionophores remain a latent therapeutic avenue to address both the global burden of antimicrobial resistance, and the unmet clinical need for new antimicrobial therapies. This review will provide an overview of the broad-spectrum antimicrobial and immunomodulatory properties of ionophores, and their potential uses in clinical medicine for combatting infection.
Collapse
|
40
|
Alkhalifah DHM. Evaluation of antimicrobial activity of bacterial symbionts isolated from wild field cockroach Blattella vaga from Saudi Arabia. Saudi J Biol Sci 2021; 28:6239-6244. [PMID: 34759743 PMCID: PMC8568709 DOI: 10.1016/j.sjbs.2021.06.092] [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: 06/02/2021] [Revised: 06/21/2021] [Accepted: 06/27/2021] [Indexed: 11/27/2022] Open
Abstract
Drug-resistant pathogens form the main threat to global health during the current century. Annually, a lot of patients die in hospitals due to infection with one or more drug-resistant bacteria especially Staphylococcus aureus (MRSA). In the absence of new effective antimicrobial drugs, the number of deaths said to be increased. Searching for new antibiotics in our backyard form a part of scientist strategies to solve such serious health problem. Insects consider one of such interesting sources of the new era of antimicrobial drugs. Cockroaches as an example can live and adapt in a polluted area for a long time, so through this work field cockroach, Blattella vaga was collected from two semi-wild areas around Riyadh, Saudi Arabia for isolation of gut bacteria searching for new antimicrobial agents. Three species of bacteria were identified from field cockroach gut: Bacillus licheniformis, Bacillus subtilis, and Kocuria rosea. The three species were isolated, purified, and tested for their antimicrobial activity against four drug-resistant pathogens (three bacteria: Salmonella enterica (ATCC25566), Staphylococcus aureus (MRSA) (Clinical strain), and Streptococcus mutans (RCMB 017(1) ATCC ® 25175™) and one fungus: Candida albicans (RCMB005003(1) ATCC® 10231™)). The results show no antimicrobial activity of Bacillus subtilis and very good activity Bacillus licheniformis and Kocuria rosea. Bacillus licheniformis gives very effective activity against Candida albicans while Kocuria rosea is effective against MRSA and Streptococcus mutans. None of the gut isolated bacteria show any activity against Salmonella enterica. Such results revealed that the metabolites of these bacteria could be used as substitutes to the already used antibiotics to overcome the problem of multidrug-resistant human pathogens.
Collapse
Affiliation(s)
- Dalal Hussien M Alkhalifah
- Biology Department, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
41
|
Espinoza LC, Sosa L, Granda PC, Bozal N, Díaz-Garrido N, Chulca-Torres B, Calpena AC. Development of a Topical Amphotericin B and Bursera graveolens Essential Oil-Loaded Gel for the Treatment of Dermal Candidiasis. Pharmaceuticals (Basel) 2021; 14:ph14101033. [PMID: 34681257 PMCID: PMC8538170 DOI: 10.3390/ph14101033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 01/02/2023] Open
Abstract
The higher molecular weight and low solubility of amphotericin B (AmB) hinders its topical administration. The aim of this study was to incorporate Bursera graveolens essential oil into an AmB topical gel (AmB + BGEO gel) in order to promote the diffusion of the drug through the skin in the treatment of cutaneous candidiasis. AmB + BGEO gel formulation was determined using a factorial experiment. Physical and chemical parameters, stability, in vitro release profile and ex vivo permeation in human skin were evaluated. In vitro antimicrobial activity was studied using strains of C. albicans, C. glabrata and C. parapsilosis. The tolerability was evaluated using in vitro and in vivo models. AmB + BGEO gel presented appropriate characteristics for topical administration, including pH of 5.85, pseudoplastic behavior, optimal extensibility, as well as high stability and acceptable tolerability. In vitro release studies showed that the formulation releases the drug following a Boltzmann sigmoidal model. Finally, AmB + BGEO gel exhibited higher amount of drug retained inside the skin and lower Minimum Inhibitory Concentration than a formulation sans essential oil. Therefore, these results suggest that the incorporation of B. graveolens essential oil in the formulation could be used as strategy to promote a local effect in the treatment of cutaneous candidiasis.
Collapse
Affiliation(s)
- Lupe Carolina Espinoza
- Departamento de Química, Universidad Técnica Particular de Loja, Loja 1101608, Ecuador; (L.C.E.); (B.C.-T.)
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, 08028 Barcelona, Spain;
| | - Lilian Sosa
- Faculty of Chemical Sciences and Pharmacy, National Autonomous University of Honduras (UNAH), Tegucigalpa 11101, Honduras;
| | - Paulo C. Granda
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, 08028 Barcelona, Spain;
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
| | - Nuria Bozal
- Department of Biology, Healthcare and the Environment, School of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain;
| | - Natalia Díaz-Garrido
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain;
- Institute of Biomedicine of the University of Barcelona-Sant Joan de Déu Research Institute (IBUB-IRSJD), 08028 Barcelona, Spain
| | - Brenda Chulca-Torres
- Departamento de Química, Universidad Técnica Particular de Loja, Loja 1101608, Ecuador; (L.C.E.); (B.C.-T.)
| | - Ana Cristina Calpena
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, 08028 Barcelona, Spain;
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
- Correspondence:
| |
Collapse
|
42
|
Chen J, Hu N, Xu H, Liu Q, Yu X, Zhang Y, Huang Y, Tan J, Huang X, Zeng L. Molecular Epidemiology, Antifungal Susceptibility, and Virulence Evaluation of Candida Isolates Causing Invasive Infection in a Tertiary Care Teaching Hospital. Front Cell Infect Microbiol 2021; 11:721439. [PMID: 34604110 PMCID: PMC8479822 DOI: 10.3389/fcimb.2021.721439] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/09/2021] [Indexed: 01/08/2023] Open
Abstract
Background The incidence of invasive candidiasis is increasing worldwide. However, the epidemiology, antifungal susceptibility, and virulence of Candida spp. in most hospitals remain unclear. This study aimed to evaluate invasive candidiasis in a tertiary care hospital in Nanchang City, China. Methods MALDI-TOF MS and 18S rDNA ITS sequencing were used to identify Candida strains. Randomly amplified polymorphic DNA analysis was used for molecular typing; biofilm production, caseinase, and hemolysin activities were used to evaluate virulence. The Sensititre™ YeastOne YO10 panel was used to examine antifungal susceptibility. Mutations in ERG11 and the hotspot regions of FKS1 of drug-resistant strains were sequenced to evaluate the possible mechanisms of antifungal resistance. Results We obtained 110 Candida strains, which included 40 Candida albicans (36.36%), 37 C. parapsilosis (33.64%), 21 C. tropicalis (19.09%), 9 C. glabrata (8.18%), 2 C. rugose (1.82%), and 1 C. haemulonii (0.91%) isolates. At a limiting point of 0.80, C. albicans isolates could be grouped into five clusters, C. parapsilosis and C. tropicalis isolates into seven clusters, and C. glabrata isolates into only one cluster comprising six strains by RAPD typing. Antifungal susceptibility testing revealed that the isolates showed the greatest overall resistance against fluconazole (6.36%), followed by voriconazole (4.55%). All C. albicans and C. parapsilosis isolates exhibited 100% susceptibility to echinocandins (i.e., anidulafungin, caspofungin, and micafungin), whereas one C. glabrata strain was resistant to echinocandins. The most common amino acid substitutions noted in our study was 132aa (Y132H, Y132F) in the azole-resistant strains. No missense mutation was identified in the hotpot regions of FKS1. Comparison of the selected virulence factors detectable in a laboratory environment, such as biofilm, caseinase, and hemolysin production, revealed that most Candida isolates were caseinase and hemolysin producers with a strong activity (Pz < 0.69). Furthermore, C. parapsilosis had greater total biofilm biomass (average Abs620 = 0.712) than C. albicans (average Abs620 = 0.214, p < 0.01) or C. tropicalis (average Abs620 = 0.450, p < 0.05), although all C. glabrata strains were either low- or no-biofilm producers. The virulence level of the isolates from different specimen sources or clusters showed no obvious correlation. Interesting, 75% of the C. albicans from cluster F demonstrated azole resistance, whereas two azole-resistant C. tropicalis strains belonged to the cluster Y. Conclusion This study provides vital information regarding the epidemiology, pathogenicity, and antifungal susceptibility of Candida spp. in patients admitted to Nanchang City Hospital.
Collapse
Affiliation(s)
- Junzhu Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Niya Hu
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hongzhi Xu
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Department of Preventive Medicine and Public Health, School of Public Health, Nanchang University, Nanchang, China
| | - Qiong Liu
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Xiaomin Yu
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Yuping Zhang
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Yongcheng Huang
- Department of Preventive Medicine and Public Health, School of Public Health, Nanchang University, Nanchang, China
| | - Junjun Tan
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Xiaotian Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Lingbing Zeng
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Department of Preventive Medicine and Public Health, School of Public Health, Nanchang University, Nanchang, China
| |
Collapse
|
43
|
Fiołka MJ, Czaplewska P, Wójcik-Mieszawska S, Lewandowska A, Lewtak K, Sofińska-Chmiel W, Buchwald T. Metabolic, structural, and proteomic changes in Candida albicans cells induced by the protein-carbohydrate fraction of Dendrobaena veneta coelomic fluid. Sci Rep 2021; 11:16711. [PMID: 34408181 PMCID: PMC8373886 DOI: 10.1038/s41598-021-96093-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 08/02/2021] [Indexed: 01/14/2023] Open
Abstract
The isolated protein-polysaccharide fraction (AAF) from the coelomic fluid of Dendrobaena veneta earthworm shows effective activity against Candida albicans yeast. Fungal cells of the clinical strain after incubation with the active fraction were characterized by disturbed cell division and different morphological forms due to the inability to separate the cells from each other. Staining of the cells with acridine orange revealed a change in the pH of the AAF-treated cells. It was observed that, after the AAF treatment, the mitochondrial DNA migrated towards the nuclear DNA, whereupon both merged into a single nuclear structure, which preceded the apoptotic process. Cells with a large nucleus were imaged with the scanning electron cryomicroscopy (Cryo-SEM) technique, while enlarged mitochondria and the degeneration of cell structures were shown by transmission electron microscopy (TEM). The loss of the correct cell shape and cell wall integrity was visualized by both the TEM and SEM techniques. Mass spectrometry and relative quantitative SWATH MS analysis were used to determine the reaction of the C. albicans proteome to the components of the AAF fraction. AAF was observed to influence the expression of mitochondrial and oxidative stress proteins. The oxidative stress in C. albicans cells caused by the action of AAF was demonstrated by fluorescence microscopy, proteomic methods, and XPS spectroscopy. The secondary structure of AAF proteins was characterized by Raman spectroscopy. Analysis of the elemental composition of AAF confirmed the homogeneity of the preparation. The observed action of AAF, which targets not only the cell wall but also the mitochondria, makes the preparation a potential antifungal drug killing the cells of the C. albicans pathogen through apoptosis.
Collapse
Affiliation(s)
- Marta J Fiołka
- Department of Immunobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033, Lublin, Poland.
| | - Paulina Czaplewska
- Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Sylwia Wójcik-Mieszawska
- Department of Immunobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033, Lublin, Poland
| | - Aleksandra Lewandowska
- Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Kinga Lewtak
- Department of Cell Biology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland
| | - Weronika Sofińska-Chmiel
- Analytical Laboratory, Institute of Chemical Sciences, Maria Curie-Skłodowska University, Lublin, Poland
| | - Tomasz Buchwald
- Faculty of Materials Science and Technical Physics, Institute of Materials Research and Quantum Engineering, Poznan University of Technology, Poznań, Poland
| |
Collapse
|
44
|
Musumeci S, Coen M, Leidi A, Schrenzel J. The human gut mycobiome and the specific role of Candida albicans: where do we stand, as clinicians? Clin Microbiol Infect 2021; 28:58-63. [PMID: 34363944 DOI: 10.1016/j.cmi.2021.07.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/08/2021] [Accepted: 07/22/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND The so-called 'mycobiome' has progressively acquired interest and increased the complexity of our understanding of the human gut microbiota. Several questions are arising concerning the role of fungi (and in particular of Candida albicans), the so-called 'mycobiome', that has been neglected for a long time and only recently gained interest within the scientific community. There is no consensus on mycobiome normobiosis because of its instability and variability. This review aims to raise awareness about this interesting topic and provide a framework to guide physicians faced with such questions. OBJECTIVES To summarize current knowledge and discuss current and potential implications of the mycobiome in clinical practice. SOURCES We performed a review of the existing literature in Medline Pubmed. CONTENT This review identifies several studies showing associations between specific mycobiome profiles and health. Fungi represent a significant biomass within the microbiota and several factors, such as diet, sex, age, co-morbidities, medications, immune status and inter-kingdom interactions, can influence its structure and population. The human gut mycobiota is indeed a key factor for several physiological processes (e.g. training of the immune system against infections) and pathological processes (e.g. immunological/inflammatory disorders, inflammatory bowel diseases, metabolic syndromes). Moreover, the mycobiome (and C. albicans in particular) could influence an even broader spectrum of conditions such as psychiatric diseases (depression, schizophrenia, bipolar disorder) or chronic viral infections (human immunodeficiency virus, hepatitis B virus); moreover, it could be implicated in tumorigenesis. IMPLICATIONS Candida albicans is a well-known opportunistic pathogen and a major component of the mycobiome but its role in the gastrointestinal tract is still poorly understood. From a potential screening biomarker to a key factor for several pathological processes, its presence could influence or even modify our clinical practice.
Collapse
Affiliation(s)
- Stefano Musumeci
- Service of Internal Medicine, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Matteo Coen
- Service of Internal Medicine, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland; Unit of Development and Research in Medical Education (UDREM), Faculty of Medicine, Geneva, Switzerland.
| | - Antonio Leidi
- Service of Internal Medicine, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Jacques Schrenzel
- Bacteriology Laboratory, Department of Diagnostics, Geneva University Hospitals, Geneva, Switzerland; Genomic Research Laboratory, Department of Medicine, Geneva University Hospitals and University of Geneva, Geneva, Switzerland; Division of Infectious Diseases, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
45
|
Oral Candidosis: Pathophysiology and Best Practice for Diagnosis, Classification, and Successful Management. J Fungi (Basel) 2021; 7:jof7070555. [PMID: 34356934 PMCID: PMC8306613 DOI: 10.3390/jof7070555] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/10/2021] [Accepted: 07/11/2021] [Indexed: 01/12/2023] Open
Abstract
Oral candidosis is the most common fungal infection that frequently occurs in patients debilitated by other diseases or conditions. No candidosis happens without a cause; hence oral candidosis has been branded as a disease of the diseased. Prior research has identified oral candidosis as a mark of systemic diseases, such as hematinic deficiency, diabetes mellitus, leukopenia, HIV/AIDS, malignancies, and carbohydrate-rich diet, drugs, or immunosuppressive conditions. An array of interaction between Candida and the host is dynamic and complex. Candida exhibits multifaceted strategies for growth, proliferation, evasion of host defenses, and survival within the host to induce fungal infection. Oral candidosis presents a variety of clinical forms, including pseudomembranous candidosis, erythematous candidosis, angular cheilitis, median rhomboid glossitis, cheilocandidosis, juxtavermillion candidosis, mucocutaneous candidosis, hyperplastic candidosis, oropharyngeal candidosis, and rare suppurative candidosis. The prognosis is usually favorable, but treatment failure or recurrence is common due to either incorrect diagnosis, missing other pathology, inability to address underlying risk factors, or inaccurate prescription of antifungal agents. In immunocompromised patients, oropharyngeal candidosis can spread to the bloodstream or upper gastrointestinal tract, leading to potentially lethal systemic candidosis. This review therefore describes oral candidosis with regard to its pathophysiology and best practice for diagnosis, practical classification, and successful management.
Collapse
|
46
|
Antifungal Azoles as Tetracycline Resistance Modifiers in Staphylococcus aureus. Appl Environ Microbiol 2021; 87:e0015521. [PMID: 33990311 DOI: 10.1128/aem.00155-21] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus aureus has developed resistance to antimicrobials since their first use. The S. aureus major facilitator superfamily (MFS) efflux pump Tet(K) contributes to resistance to tetracyclines. The efflux pump diminishes antibiotic accumulation, and biofilm hampers the diffusion of antibiotics. None of the currently known compounds have been approved as efflux pump inhibitors (EPIs) for clinical use. In the current study, we screened clinically approved drugs for possible Tet(K) efflux pump inhibition. By performing in silico docking followed by in vitro checkerboard assays, we identified five azoles (the fungal ergosterol synthesis inhibitors) showing putative EPI-like potential with a fractional inhibitory concentration index of ≤0.5, indicating synergism. The functionality of the azoles was confirmed using ethidium bromide (EtBr) accumulation and efflux inhibition assays. In time-kill kinetics, the combination treatment with butoconazole engendered a marked increase in the bactericidal capacity of tetracycline. When assessing the off-target effects of the azoles, we observed no disruption of bacterial membrane permeability and polarization. Finally, the combination of azoles with tetracycline led to a significant eradication of preformed mature biofilms. This study demonstrates that azoles can be repurposed as putative Tet(K) EPIs and to reduce biofilm formation at clinically relevant concentrations. IMPORTANCE Staphylococcus aureus uses efflux pumps to transport antibiotics out of the cell and thus increases the dosage at which it endures antibiotics. Also, efflux pumps play a role in biofilm formation by the excretion of extracellular matrix molecules. One way to combat these pathogens may be to reduce the activity of efflux pumps and thereby increase pathogen sensitivity to existing antibiotics. We describe the in silico-based screen of clinically approved drugs that identified antifungal azoles inhibiting Tet(K), a pump that belongs to the major facilitator superfamily, and showed that these compounds bind to and block the activity of the Tet(K) pump. Azoles enhanced the susceptibility of tetracycline against S. aureus and its methicillin-resistant strains. The combination of azoles with tetracycline led to a significant reduction in preformed biofilms. Repurposing approved drugs may help solve the classical toxicity issues related to efflux pump inhibitors.
Collapse
|
47
|
Ren T, Zhu H, Tian L, Yu Q, Li M. Candida albicans infection disturbs the redox homeostasis system and induces reactive oxygen species accumulation for epithelial cell death. FEMS Yeast Res 2021; 20:5643898. [PMID: 31769804 DOI: 10.1093/femsyr/foz081] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 11/25/2019] [Indexed: 02/07/2023] Open
Abstract
Candida albicans is a common pathogenic fungus with high mortality in immunocompromised patients. However, the mechanism by which C. albicans invades host epithelial cells and causes serious tissue damage remains to be further investigated. In this study, we established the C. albicans-293T renal epithelial cell interaction model to investigate the mechanism of epithelial infection by this pathogen. It was found that C. albicans infection causes severe cell death and reactive oxygen species (ROS) accumulation in epithelial cells. Further investigations revealed that C. albicans infection might up-regulate expression of nicotinamide adenine dinucleotide phosphate (NAPDH) oxidase (NOX), inhibit the activity of the antioxidant enzymes superoxide dismutase (SOD) and catalase (CAT), and suppress the p38-Nrf2-heme oxygenase-1 (HO-1) pathway which plays an important role in the elimination of intracellular ROS. Furthermore, epithelial cell death caused by the fungal infection could be strikingly alleviated by addition of the antioxidant agent glutathione, indicating the critical role of ROS accumulation in cell death caused by the fungus. This study revealed that disturbance of the redox homeostasis system and ROS accumulation in epithelial cells is involved in cell death caused by C. albicans infection, which sheds light on the application of antioxidants in the suppression of tissue damage caused by fungal infection.
Collapse
Affiliation(s)
- Tongtong Ren
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Science, Nankai University, Tianjin 300071, P. R. China
| | - Hangqi Zhu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Science, Nankai University, Tianjin 300071, P. R. China
| | - Lei Tian
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Science, Nankai University, Tianjin 300071, P. R. China
| | - Qilin Yu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Science, Nankai University, Tianjin 300071, P. R. China
| | - Mingchun Li
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Science, Nankai University, Tianjin 300071, P. R. China
| |
Collapse
|
48
|
Price A, Morris TC, White HA, Hamilton RA. Cutaneous phototoxic reaction to intravenous micafungin in the outpatient setting: A case report. CLINICAL INFECTION IN PRACTICE 2021. [DOI: 10.1016/j.clinpr.2020.100061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
49
|
Dennyson Savariraj A, Salih A, Alam F, Elsherif M, AlQattan B, Khan AA, Yetisen AK, Butt H. Ophthalmic Sensors and Drug Delivery. ACS Sens 2021; 6:2046-2076. [PMID: 34043907 PMCID: PMC8294612 DOI: 10.1021/acssensors.1c00370] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 05/17/2021] [Indexed: 12/15/2022]
Abstract
Advances in multifunctional materials and technologies have allowed contact lenses to serve as wearable devices for continuous monitoring of physiological parameters and delivering drugs for ocular diseases. Since the tear fluids comprise a library of biomarkers, direct measurement of different parameters such as concentration of glucose, urea, proteins, nitrite, and chloride ions, intraocular pressure (IOP), corneal temperature, and pH can be carried out non-invasively using contact lens sensors. Microfluidic contact lens sensor based colorimetric sensing and liquid control mechanisms enable the wearers to perform self-examinations at home using smartphones. Furthermore, drug-laden contact lenses have emerged as delivery platforms using a low dosage of drugs with extended residence time and increased ocular bioavailability. This review provides an overview of contact lenses for ocular diagnostics and drug delivery applications. The designs, working principles, and sensing mechanisms of sensors and drug delivery systems are reviewed. The potential applications of contact lenses in point-of-care diagnostics and personalized medicine, along with the significance of integrating multiplexed sensing units together with drug delivery systems, have also been discussed.
Collapse
Affiliation(s)
| | - Ahmed Salih
- Department
of Mechanical Engineering, Khalifa University
of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Fahad Alam
- Department
of Mechanical Engineering, Khalifa University
of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Mohamed Elsherif
- Department
of Mechanical Engineering, Khalifa University
of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Bader AlQattan
- Department
of Mechanical Engineering, Khalifa University
of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Ammar A. Khan
- Department
of Chemical Engineering, Imperial College
London, London SW7 2AZ, United Kingdom
| | - Ali K. Yetisen
- Department
of Physics, Lahore University of Management
Sciences, Lahore Cantonment 54792, Lahore, Pakistan
| | - Haider Butt
- Department
of Mechanical Engineering, Khalifa University
of Science and Technology, Abu Dhabi, United Arab Emirates
| |
Collapse
|
50
|
Photodynamic Therapy Combined with Antibiotics or Antifungals against Microorganisms That Cause Skin and Soft Tissue Infections: A Planktonic and Biofilm Approach to Overcome Resistances. Pharmaceuticals (Basel) 2021; 14:ph14070603. [PMID: 34201530 PMCID: PMC8308592 DOI: 10.3390/ph14070603] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 12/18/2022] Open
Abstract
The present review covers combination approaches of antimicrobial photodynamic therapy (aPDT) plus antibiotics or antifungals to attack bacteria and fungi in vitro (both planktonic and biofilm forms) focused on those microorganisms that cause infections in skin and soft tissues. The combination can prevent failure in the fight against these microorganisms: antimicrobial drugs can increase the susceptibility of microorganisms to aPDT and prevent the possibility of regrowth of those that were not inactivated during the irradiation; meanwhile, aPDT is effective regardless of the resistance pattern of the strain and their use does not contribute to the selection of antimicrobial resistance. Additive or synergistic antimicrobial effects in vitro are evaluated and the best combinations are presented. The use of combined treatment of aPDT with antimicrobials could help overcome the difficulty of fighting high level of resistance microorganisms and, as it is a multi-target approach, it could make the selection of resistant microorganisms more difficult.
Collapse
|