1
|
Arévalo-Jaimes BV, Admella J, Torrents E. Who arrived first? Priority effects on Candida albicans and Pseudomonas aeruginosa dual biofilms. Commun Biol 2025; 8:160. [PMID: 39901054 PMCID: PMC11790929 DOI: 10.1038/s42003-025-07609-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 01/27/2025] [Indexed: 02/05/2025] Open
Abstract
Historical processes in community assembly, such as species arrival order, influence interactions, causing priority effects. Candida albicans and Pseudomonas aeruginosa often co-occur in biofilm-based infections of the skin, lungs, and medical devices. Their predominantly antagonistic relationship involves complex physical and chemical interactions. However, the presence and implications of priority effects among these microorganisms remain largely unexplored. Here, we investigate the presence and impact of priority effect in dual-species biofilms using clinical isolates. By varying inoculation order, we observe significant changes in biofilm composition, structure, virulence, and antimicrobial susceptibility. The first colonizer has an advantage for surface colonization. Consecutive colonization increases biofilm virulence and negates C. albicans' protective effect on P. aeruginosa PAET1 against meropenem treatment. Finally, we propose N-acetylcysteine as an adjuvant for treating C. albicans and P. aeruginosa interkingdom infections, working independently of priority effects.
Collapse
Affiliation(s)
- Betsy V Arévalo-Jaimes
- Bacterial infections and antimicrobial therapies group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Joana Admella
- Bacterial infections and antimicrobial therapies group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Eduard Torrents
- Bacterial infections and antimicrobial therapies group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain.
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
2
|
da Silva Oliveira DD, Sousa Silva NB, Jacques Dit Lapierre TJW, Lemes de Souza S, Ferreira Brito NP, Cassiano Martinho AC, Clemente Dias RF, Nascimento Farago D, Michelan-Duarte S, Consolin Chelucci R, de Moura Lodi Cruz MGF, de Melo Resende D, Andricopulo AD, Fonseca Murta SM, Ferreira LLG, Gomes Martins CH, de Oliveira Rezende Júnior C. Discovery of Arylpiperazines with Broad-Spectrum Antimicrobial Activity and Favorable Pharmacokinetic Profiles. Chem Biodivers 2025; 22:e202402100. [PMID: 39327235 DOI: 10.1002/cbdv.202402100] [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: 08/24/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 09/28/2024]
Abstract
Microorganisms can induce diseases with significant clinical implications for human health. Multidrug-resistant microorganisms have been on the rise worldwide over the past few decades, and no new antibiotics have been introduced to the market in a considerable amount of time. Such situation highlights the urgency of discovering new antimicrobial drugs to address this pressing issue. Therefore, the objective of this study was to identify bioactive compounds against 15 species of bacteria and 5 species of fungi of clinical relevance through in vitro screening of 58 synthetic compounds from four chemical classes of our internal library of synthetic compounds. Our findings highlight arylpiperazines 18, 20, 26, 27, and 29, and the aminothiazole 50, as potent broad-spectrum antimicrobials (MICs=12.5-15.6 μg mL-1) against clinically relevant bacteria and fungi. Additionally, these compounds displayed low cytotoxicity against various host cells and a favorable in vitro pharmacokinetic profile for oral administration. Indeed, all six showed adequate lipophilicity, high gastrointestinal permeability, metabolic stability in human and mouse liver microsomes, and satisfactory aqueous solubility. Thus, they emerge as promising starting points for hit-to-lead studies towards new antibacterial and antifungal agents, especially against Staphylococcus epidermidis, Staphylococcus aureus, Lactobacillus paracasei and Candida orthopsilosis.
Collapse
Affiliation(s)
- Douglas Davison da Silva Oliveira
- Laboratório de Síntese de Candidatos a Fármacos (LaSFar), Institute of Chemistry, Federal University of Uberlândia, Uberlândia-MG, 38400-902, Brazil
| | - Nagela Bernadelli Sousa Silva
- Laboratory of Antimicrobial Testing (LEA), Institute of Biomedical Sciences (ICBIM), Federal University of Uberlândia, Uberlândia-MG, 38400-902, Brazil
| | | | - Sara Lemes de Souza
- Laboratory of Antimicrobial Testing (LEA), Institute of Biomedical Sciences (ICBIM), Federal University of Uberlândia, Uberlândia-MG, 38400-902, Brazil
| | - Nícolas Peterson Ferreira Brito
- Laboratório de Síntese de Candidatos a Fármacos (LaSFar), Institute of Chemistry, Federal University of Uberlândia, Uberlândia-MG, 38400-902, Brazil
| | - Ana Clara Cassiano Martinho
- Laboratório de Síntese de Candidatos a Fármacos (LaSFar), Institute of Chemistry, Federal University of Uberlândia, Uberlândia-MG, 38400-902, Brazil
| | - Renieidy Flávia Clemente Dias
- Laboratório de Síntese de Candidatos a Fármacos (LaSFar), Institute of Chemistry, Federal University of Uberlândia, Uberlândia-MG, 38400-902, Brazil
| | - Danilo Nascimento Farago
- Laboratório de Síntese de Candidatos a Fármacos (LaSFar), Institute of Chemistry, Federal University of Uberlândia, Uberlândia-MG, 38400-902, Brazil
| | - Simone Michelan-Duarte
- Laboratório de Química Medicinal e Computacional (LQMC), Instituto de Física de São Carlos (IFSC), Universidade de São Paulo (USP), São Carlos-SP, 13563-120, Brazil
| | - Rafael Consolin Chelucci
- Laboratório de Química Medicinal e Computacional (LQMC), Instituto de Física de São Carlos (IFSC), Universidade de São Paulo (USP), São Carlos-SP, 13563-120, Brazil
| | | | - Daniela de Melo Resende
- Grupo de Genômica Funcional de Parasitos, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ Minas), Belo Horizonte-MG, 30190-002, Brazil
| | - Adriano D Andricopulo
- Laboratório de Química Medicinal e Computacional (LQMC), Instituto de Física de São Carlos (IFSC), Universidade de São Paulo (USP), São Carlos-SP, 13563-120, Brazil
| | - Silvane Maria Fonseca Murta
- Grupo de Genômica Funcional de Parasitos, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ Minas), Belo Horizonte-MG, 30190-002, Brazil
| | - Leonardo L G Ferreira
- Laboratório de Química Medicinal e Computacional (LQMC), Instituto de Física de São Carlos (IFSC), Universidade de São Paulo (USP), São Carlos-SP, 13563-120, Brazil
| | - Carlos Henrique Gomes Martins
- Laboratory of Antimicrobial Testing (LEA), Institute of Biomedical Sciences (ICBIM), Federal University of Uberlândia, Uberlândia-MG, 38400-902, Brazil
| | - Celso de Oliveira Rezende Júnior
- Laboratório de Síntese de Candidatos a Fármacos (LaSFar), Institute of Chemistry, Federal University of Uberlândia, Uberlândia-MG, 38400-902, Brazil
| |
Collapse
|
3
|
Babady NE, Chiu CY, Craney A, Gaston DC, Hicklen RS, Hogan CA, John TM, Stewart AG. Diagnosis and management of invasive fungal diseases by next-generation sequencing: are we there yet? Expert Rev Mol Diagn 2024:1-14. [PMID: 39623670 DOI: 10.1080/14737159.2024.2436396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 11/27/2024] [Indexed: 12/13/2024]
Abstract
INTRODUCTION Invasive fungal diseases (IFDs) are a serious threat to immunocompromised patients. Routine diagnostic methods have limited performance in identifying IFDs. Next-generation sequencing (NGS), including metagenomic NGS (mNGS) and whole-genome sequencing (WGS), recently emerged as diagnostic methods that could provide more accurate and timely diagnoses and management of IFDs. AREAS COVERED This article describes the emergence of NGS as a diagnostic tool to address the limitations of current tests. The literature regarding its application and clinical utility in the diagnosis of IFDs is reviewed. Practical considerations, challenges, and opportunities as they relate to the development and implementation of mNGS and WGS for fungal pathogens are discussed. EXPERT OPINION NGS emerged over a decade ago with the potential to solve many of the challenges in diagnosing infectious diseases, including IFDs. However, published literature has yielded conflicting data about its clinical utility. The increased clinical adoption of NGS is improving our understanding of how to interpret and use its results to guide actionable decisions. Still, several gaps remain. As the cost, effort, and expertise involved in performing NGS decrease and the reporting of its results becomes standardized, NGS is poised to fill current gaps in the diagnosis of IFDs.
Collapse
Affiliation(s)
- N Esther Babady
- Clinical Microbiology Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Charles Y Chiu
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
- Department of Medicine, Division of Infectious Diseases, University of California, San Francisco, CA, USA
| | | | - David C Gaston
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rachel S Hicklen
- Research Medical Library, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Catherine A Hogan
- British Columbia Centre for Disease Control, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Teny M John
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Adam G Stewart
- Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane and Women's Hospital Campus, Brisbane, Australia
- Central Microbiology, Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, Australia
| |
Collapse
|
4
|
Haley E, Luke N, Korman H, Rao GS, Baunoch D, Chen X, Havrilla J, Mathur M. Comparing Prescribing Behaviors and Clinician Experiences Between Multiplex PCR/Pooled Antibiotic Susceptibility Testing and Standard Urine Culture in Complicated UTI Cases. J Clin Med 2024; 13:7453. [PMID: 39685910 DOI: 10.3390/jcm13237453] [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: 10/02/2024] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Background/Objectives: We aimed to compare the prescribing behavior and clinical experience of urology providers when using the combined multiplex polymerase chain reaction (M-PCR)/Pooled Antibiotic Susceptibility Testing (P-AST) diagnostic test versus the standard urine culture (SUC) in the same set of patients previously reported to have improved clinical outcomes with M-PCR/P-AST. Methods: We conducted a multi-centered, prospective, observational study (clinical trial registration: NCT05091931) with Western Institutional Review Board (IRB) approval (20214705). Adult subjects were split between the M-PCR/P-AST (n = 250) and SUC arms (n = 135). Treatment details were determined by clinician and subject surveys. Differences in prescribed antibiotics were compared using the Chi-square or Fisher's exact test. Results: There was no significant difference in the overall use of "access" antibiotics (p = 1.0) or first-line drugs (p = 0.4483) between M-PCR/P-AST and SUC. Nitrofurantoin (p = 0.0172) and metronidazole (p = 0.0309) were more frequently used with M-PCR/P-AST, while amoxicillin/clavulanate (p = 0.0008), cefuroxime (p = 0.0378), and ertapenem (p = 0.0378) were more frequently used with SUC. Conclusions: The use of M-PCR/P-AST to guide complicated UTI management was not associated with the increased use of non-first-line antibiotics, such as carbapenems, compared to SUC. Combined with the prior reported evidence of improved patient outcomes in this same set of patients, this test should be considered for utilization when managing complicated UTI cases.
Collapse
Affiliation(s)
- Emery Haley
- Department of Clinical Research, Pathnostics, Irvine, CA 92618, USA
| | - Natalie Luke
- Department of Clinical Research, Pathnostics, Irvine, CA 92618, USA
| | - Howard Korman
- School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | | | - David Baunoch
- Department of Research and Development, Pathnostics, Irvine, CA 92618, USA
| | - Xiaofei Chen
- Department of Informatics, Pathnostics, Irvine, CA 92618, USA
| | - Jim Havrilla
- Department of Informatics, Pathnostics, Irvine, CA 92618, USA
| | - Mohit Mathur
- Department of Medical Affairs, Pathnostics, Irvine, CA 92618, USA
| |
Collapse
|
5
|
Hacioglu M, Yilmaz FN, Yetke HI, Haciosmanoglu-Aldogan E. Synergistic effects of quorum-sensing molecules and antimicrobials against Candida albicans and Pseudomonas aeruginosa biofilms: in vitro and in vivo studies. J Antimicrob Chemother 2024; 79:2828-2836. [PMID: 39212125 DOI: 10.1093/jac/dkae293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Candida albicans can form polymicrobial biofilms with other microorganisms, such as Pseudomonas aeruginosa, at infection sites. OBJECTIVES As biofilms are highly resistant to antibiotics there is a need for new antibiofilm agents that have unique targets and modes of action. METHODS In this study the antibiofilm effects of two quorum-sensing molecules (QSMs), farnesol and tyrosol, were investigated alone and in combination with antibiotics (aztreonam, colistin, tobramycin) and antifungals (fluconazole, amphotericin B, caspofungin), against single- and dual-species biofilms of C. albicans and P. aeruginosa in in vitro and in vivo systems. RESULTS It was observed that QSMs alone, especially farnesol, showed at least a 1-log reduction against preformed single- and dual-species biofilms of C. albicans and P. aeruginosa. Combination of QSMs with colistin or fluconazole was found to be effective against both single- and dual-species biofilms in vitro. Increased survival was observed in C. elegans when treated with colistin or fluconazole in combination with QSMs, compared with no treatment. Additionally, the QSMs and colistin and farnesol combinations effectively inhibited biofilm formation by C. albicans and P. aeruginosa on bronchial epithelial cells, and reduced IL-1β expression in lung bronchial epithelial cells. CONCLUSIONS There is a need for effective treatments for bacterial-fungal biofilm infections and, to our knowledge, there have been no studies of QSMs and antimicrobial combinations against dual-species biofilms involving C. albicans and P. aeruginosa. Hence these findings will make a significant contribution to the literature.
Collapse
Affiliation(s)
- Mayram Hacioglu
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Istanbul University, Beyazit, Istanbul 34116, Türkiye
| | - Fatima Nur Yilmaz
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Istanbul University, Beyazit, Istanbul 34116, Türkiye
| | - Hande Ipek Yetke
- Department of Biophysics, Faculty of Medicine, Bezmialem Vakif University, Vatan Street Fatih, Istanbul 34098, Türkiye
| | - Ebru Haciosmanoglu-Aldogan
- Department of Biophysics, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Fatih, Istanbul 34093, Türkiye
| |
Collapse
|
6
|
Haley E, Luke N. From Awareness to Action: Pioneering Solutions for Women's UTI Challenges in the Era of Precision Medicine. Int J Womens Health 2024; 16:1595-1605. [PMID: 39359902 PMCID: PMC11446210 DOI: 10.2147/ijwh.s477476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024] Open
Abstract
This article aims to bring clinicians' awareness to the widespread impact of urinary tract infection (UTI) on the lives of women and to the advances that offer hope for future improvements in the diagnosis and management of UTI. Thanks to physiological, anatomical, and lifestyle factor differences, women face heightened vulnerability to UTIs compared to men. In fact, women are four times more likely than men to develop a UTI and around half of these women encounter UTI recurrence, which is a significant source of both physical and psychosocial burdens. Despite the current shortcomings in diagnosis and management, emerging diagnostic technologies promise to identify UTIs more accurately and rapidly, offering women hope for a revolution in UTI management. Meanwhile, clinicians have the opportunity to reduce the psychosocial burden by recognizing the value of patients' lived experiences and ensuring their care plan is in alignment with their patients' goals and expectations for medical care.
Collapse
Affiliation(s)
- Emery Haley
- Department of Clinical Research, Pathnostics, Irvine, CA, USA
| | - Natalie Luke
- Department of Clinical Research, Pathnostics, Irvine, CA, USA
| |
Collapse
|
7
|
Chakraborty A, Bandyopadhaya A, Singh VK, Kovacic F, Cha S, Oldham WM, Tzika AA, Rahme LG. The bacterial quorum sensing signal 2'-aminoacetophenone rewires immune cell bioenergetics through the Ppargc1a/Esrra axis to mediate tolerance to infection. eLife 2024; 13:RP97568. [PMID: 39269443 PMCID: PMC11398867 DOI: 10.7554/elife.97568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024] Open
Abstract
How bacterial pathogens exploit host metabolism to promote immune tolerance and persist in infected hosts remains elusive. To achieve this, we show that Pseudomonas aeruginosa (PA), a recalcitrant pathogen, utilizes the quorum sensing (QS) signal 2'-aminoacetophenone (2-AA). Here, we unveil how 2-AA-driven immune tolerization causes distinct metabolic perturbations in murine macrophages' mitochondrial respiration and bioenergetics. We present evidence indicating that these effects stem from decreased pyruvate transport into mitochondria. This reduction is attributed to decreased expression of the mitochondrial pyruvate carrier (Mpc1), which is mediated by diminished expression and nuclear presence of its transcriptional regulator, estrogen-related nuclear receptor alpha (Esrra). Consequently, Esrra exhibits weakened binding to the Mpc1 promoter. This outcome arises from the impaired interaction between Esrra and the peroxisome proliferator-activated receptor gamma coactivator 1-alpha (Ppargc1a). Ultimately, this cascade results in diminished pyruvate influx into mitochondria and, consequently reduced ATP production in tolerized murine and human macrophages. Exogenously added ATP in infected macrophages restores the transcript levels of Mpc1 and Esrra and enhances cytokine production and intracellular bacterial clearance. Consistent with the in vitro findings, murine infection studies corroborate the 2-AA-mediated long-lasting decrease in ATP and acetyl-CoA and its association with PA persistence, further supporting this QS signaling molecule as the culprit of the host bioenergetic alterations and PA persistence. These findings unveil 2-AA as a modulator of cellular immunometabolism and reveal an unprecedented mechanism of host tolerance to infection involving the Ppargc1a/Esrra axis in its influence on Mpc1/OXPHOS-dependent energy production and PA clearance. These paradigmatic findings pave the way for developing treatments to bolster host resilience to pathogen-induced damage. Given that QS is a common characteristic of prokaryotes, it is likely that 2-AA-like molecules with similar functions may be present in other pathogens.
Collapse
Affiliation(s)
- Arijit Chakraborty
- Department of Surgery, Massachusetts General Hospital, and Harvard Medical SchoolBostonUnited States
- Shriners Hospitals for Children BostonBostonUnited States
- Department of Microbiology, Harvard Medical SchoolBostonUnited States
| | - Arunava Bandyopadhaya
- Department of Surgery, Massachusetts General Hospital, and Harvard Medical SchoolBostonUnited States
- Shriners Hospitals for Children BostonBostonUnited States
| | - Vijay K Singh
- Department of Surgery, Massachusetts General Hospital, and Harvard Medical SchoolBostonUnited States
- Shriners Hospitals for Children BostonBostonUnited States
| | - Filip Kovacic
- Department of Surgery, Massachusetts General Hospital, and Harvard Medical SchoolBostonUnited States
- Department of Microbiology, Harvard Medical SchoolBostonUnited States
- Institute of Molecular Enzyme Technology, Heinrich Heine University DüsseldorfJülichGermany
| | - Sujin Cha
- Department of Surgery, Massachusetts General Hospital, and Harvard Medical SchoolBostonUnited States
| | - William M Oldham
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical SchoolBostonUnited States
| | - A Aria Tzika
- Department of Surgery, Massachusetts General Hospital, and Harvard Medical SchoolBostonUnited States
- Shriners Hospitals for Children BostonBostonUnited States
| | - Laurence G Rahme
- Department of Surgery, Massachusetts General Hospital, and Harvard Medical SchoolBostonUnited States
- Shriners Hospitals for Children BostonBostonUnited States
- Department of Microbiology, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
8
|
Chakraborty A, Bandyopadhaya A, Singh V, Kovacic F, Cha S, Oldham W, Tzika AA, Rahme L. The Bacterial Quorum-Sensing Signal 2-Aminoacetophenone Rewires Immune Cell Bioenergetics through the PGC-1α/ERRα Axis to Mediate Tolerance to Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582124. [PMID: 38464050 PMCID: PMC10925214 DOI: 10.1101/2024.02.26.582124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
How bacterial pathogens exploit host metabolism to promote immune tolerance and persist in infected hosts remains elusive. To achieve this, we show that Pseudomonas aeruginosa (PA), a recalcitrant pathogen, utilizes the quorum sensing (QS) signal 2-aminoacetophenone (2-AA). Here, we unveil how 2-AA-driven immune tolerization causes distinct metabolic perturbations in macrophages mitochondrial respiration and bioenergetics. We present evidence indicating that these effects stem from decreased pyruvate transport into mitochondria. This reduction is attributed to decreased expression of the mitochondrial pyruvate carrier (MPC1), which is mediated by diminished expression and nuclear presence of its transcriptional regulator, estrogen-related nuclear receptor alpha (ERRα). Consequently, ERRα exhibits weakened binding to the MPC1 promoter. This outcome arises from the impaired interaction between ERRα and the peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α). Ultimately, this cascade results in diminished pyruvate influx into mitochondria and, consequently reduced ATP production in tolerized macrophages. Exogenously added ATP in infected macrophages restores the transcript levels of MPC1 and ERRα and enhances cytokine production and intracellular bacterial clearance. Consistent with the in vitro findings, murine infection studies corroborate the 2-AA-mediated long-lasting decrease in ATP and acetyl-CoA and its association with PA persistence, further supporting this QS signaling molecule as the culprit of the host bioenergetic alterations and PA persistence. These findings unveil 2-AA as a modulator of cellular immunometabolism and reveal an unprecedented mechanism of host tolerance to infection involving the PGC-1α/ERRα axis in its influence on MPC1/OXPHOS-dependent energy production and PA clearance. These paradigmatic findings pave the way for developing treatments to bolster host resilience to pathogen-induced damage. Given that QS is a common characteristic of prokaryotes, it is likely that 2-AA-like molecules with similar functions may be present in other pathogens.
Collapse
|
9
|
Zhang Y, Zhang H, Xu T, Zeng L, Liu F, Huang X, Liu Q. Interactions among microorganisms open up a new world for anti-infectious therapy. FEBS J 2024; 291:1615-1631. [PMID: 36527169 DOI: 10.1111/febs.16705] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 11/12/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
The human microbiome, containing bacteria, fungi, and viruses, is a community that coexists peacefully with humans most of the time, but with the potential to cause disease under certain conditions. When the environment changes or certain stimuli are received, microbes may interact with each other, causing or increasing the severity of disease in a host. With the appropriate methods, we can make these microbiota work for us, creating new applications for human health. This review discusses the wide range of interactions between microorganisms that result in an increase in susceptibility to, severity of, and mortality of diseases, and also briefly introduces how microorganisms interact with each other directly or indirectly. The study of microbial interactions and their mechanisms has revealed a new world of treatments for infectious disease. The regulation of the balance between intestinal flora, the correct application of probiotics, and the development of effective drugs by symbiosis all demonstrate the great contributions of the microbiota to human health and its powerful potential value. Consequently, the study of interactions between microorganisms plays an essential role in identifying the causes of diseases and the development of treatments.
Collapse
Affiliation(s)
- Yejia Zhang
- Department of Medical Microbiology, School of Medicine, Jiangxi Medical College, Nanchang University, China
| | - Hanchi Zhang
- Department of Medical Microbiology, School of Medicine, Jiangxi Medical College, Nanchang University, China
- The First Clinical Medical College, Nanchang University, China
| | - Tian Xu
- Department of Medical Microbiology, School of Medicine, Jiangxi Medical College, Nanchang University, China
| | - Lingbing Zeng
- Department of Medical Microbiology, School of Medicine, Jiangxi Medical College, Nanchang University, China
- The First Clinical Medical College, Nanchang University, China
| | - Fadi Liu
- The Department of Clinical Laboratory, Children's Hospital of Jiangxi Province, Nanchang, China
| | - Xiaotian Huang
- Department of Medical Microbiology, School of Medicine, Jiangxi Medical College, Nanchang University, China
| | - Qiong Liu
- Department of Medical Microbiology, School of Medicine, Jiangxi Medical College, Nanchang University, China
| |
Collapse
|
10
|
Ramage G, Borghi E, Rodrigues CF, Kean R, Williams C, Lopez-Ribot J. Our current clinical understanding of Candida biofilms: where are we two decades on? APMIS 2023; 131:636-653. [PMID: 36932821 DOI: 10.1111/apm.13310] [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: 02/27/2023] [Accepted: 03/12/2023] [Indexed: 03/19/2023]
Abstract
Clinically we have been aware of the concept of Candida biofilms for many decades, though perhaps without the formal designation. Just over 20 years ago the subject emerged on the back of progress made from the bacterial biofilms, and academic progress pace has continued to mirror the bacterial biofilm community, albeit at a decreased volume. It is apparent that Candida species have a considerable capacity to colonize surfaces and interfaces and form tenacious biofilm structures, either alone or in mixed species communities. From the oral cavity, to the respiratory and genitourinary tracts, wounds, or in and around a plethora of biomedical devices, the scope of these infections is vast. These are highly tolerant to antifungal therapies that has a measurable impact on clinical management. This review aims to provide a comprehensive overight of our current clinical understanding of where these biofilms cause infections, and we discuss existing and emerging antifungal therapies and strategies.
Collapse
Affiliation(s)
- Gordon Ramage
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
- Study Group for Biofilms (ESGB), European Society for Clinical Microbiology and Infectious Disease, Basel, Switzerland
| | - Elisa Borghi
- Study Group for Biofilms (ESGB), European Society for Clinical Microbiology and Infectious Disease, Basel, Switzerland
- Department of Health Sciences, San Paolo Medical School, Università Degli Studi di Milano, Milan, Italy
| | - Célia Fortuna Rodrigues
- Study Group for Biofilms (ESGB), European Society for Clinical Microbiology and Infectious Disease, Basel, Switzerland
- LEPABE-Department of Chemical Engineering, Faculty of Engineering, Cooperativa de Ensino Superior Politécnico e Universitário-CESPU, Gandra, Portugal
- ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, Cooperativa de Ensino Superior Politécnico e Universitário-CESPU, Gandra, Portugal
- TOXRUN-Toxicology Research Unit, Cooperativa de Ensino Superior Politécnico e Universitário-CESPU, Gandra, Portugal
| | - Ryan Kean
- Study Group for Biofilms (ESGB), European Society for Clinical Microbiology and Infectious Disease, Basel, Switzerland
- Department of Biological Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Craig Williams
- Study Group for Biofilms (ESGB), European Society for Clinical Microbiology and Infectious Disease, Basel, Switzerland
- Microbiology Department, Morecambe Bay NHS Trust, Lancaster, UK
| | - Jose Lopez-Ribot
- Department of Biology and the South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
11
|
Khan F, Jeong GJ, Javaid A, Thuy Nguyen Pham D, Tabassum N, Kim YM. Surface adherence and vacuolar internalization of bacterial pathogens to the Candida spp. cells: Mechanism of persistence and propagation. J Adv Res 2023; 53:115-136. [PMID: 36572338 PMCID: PMC10658324 DOI: 10.1016/j.jare.2022.12.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/17/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The co-existence of Candida albicans with the bacteria in the host tissues and organs displays interactions at competitive, antagonistic, and synergistic levels. Several pathogenic bacteria take advantage of such types of interaction for their survival and proliferation. The chemical interaction involves the signaling molecules produced by the bacteria or Candida spp., whereas the physical attachment occurs by involving the surface proteins of the bacteria and Candida. In addition, bacterial pathogens have emerged to internalize inside the C. albicans vacuole, which is one of the inherent properties of the endosymbiotic relationship between the bacteria and the eukaryotic host. AIM OF REVIEW The interaction occurring by the involvement of surface protein from diverse bacterial species with Candida species has been discussed in detail in this paper. An in silico molecular docking study was performed between the surface proteins of different bacterial species and Als3P of C. albicans to explain the molecular mechanism involved in the Als3P-dependent interaction. Furthermore, in order to understand the specificity of C. albicans interaction with Als3P, the evolutionary relatedness of several bacterial surface proteins has been investigated. Furthermore, the environmental factors that influence bacterial pathogen internalization into the Candida vacuole have been addressed. Moreover, the review presented future perspectives for disrupting the cross-kingdom interaction and eradicating the endosymbiotic bacterial pathogens. KEY SCIENTIFIC CONCEPTS OF REVIEW With the involvement of cross-kingdom interactions and endosymbiotic relationships, the bacterial pathogens escape from the environmental stresses and the antimicrobial activity of the host immune system. Thus, the study of interactions between Candida and bacterial pathogens is of high clinical significance.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea.
| | - Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Aqib Javaid
- Department of Biotechnology and Bioinformatics, University of Hyderabad, India
| | - Dung Thuy Nguyen Pham
- Institute of Applied Technology and Sustainable Development, Nguyen Tat Thanh University, Ho Chi Minh City 70000, Vietnam
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Young-Mog Kim
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea; Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
12
|
Alam F, Blackburn SA, Davis J, Massar K, Correia J, Tsai HJ, Blair JMA, Hall RA. Pseudomonas aeruginosa increases the susceptibility of Candida albicans to amphotericin B in dual-species biofilms. J Antimicrob Chemother 2023; 78:2228-2241. [PMID: 37522316 PMCID: PMC10477122 DOI: 10.1093/jac/dkad228] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 07/10/2023] [Indexed: 08/01/2023] Open
Abstract
BACKGROUND Biofilms are the leading cause of nosocomial infections and are hard to eradicate due to their inherent antimicrobial resistance. Candida albicans is the leading cause of nosocomial fungal infections and is frequently co-isolated with the bacterium Pseudomonas aeruginosa from biofilms in the cystic fibrosis lung and severe burn wounds. The presence of C. albicans in multispecies biofilms is associated with enhanced antibacterial resistance, which is largely mediated through fungal extracellular carbohydrates sequestering the antibiotics. However, significantly less is known regarding the impact of polymicrobial biofilms on antifungal resistance. RESULTS Here we show that, in dual-species biofilms, P. aeruginosa enhances the susceptibility of C. albicans to amphotericin B, an effect that was biofilm specific. Transcriptional analysis combined with gene ontology enrichment analysis identified several C. albicans processes associated with oxidative stress to be differentially regulated in dual-species biofilms, suggesting that P. aeruginosa exerts oxidative stress on C. albicans, likely through the secretion of phenazines. However, the mitochondrial superoxide dismutase SOD2 was significantly down-regulated in the presence of P. aeruginosa. Monospecies biofilms of the sod2Δ mutant were more susceptible to amphotericin B, and the susceptibility of these biofilms was further enhanced by exogenous phenazines. CONCLUSIONS We propose that in dual-species biofilms, P. aeruginosa simultaneously induces mitochondrial oxidative stress, while down-regulating key detoxification enzymes, which prevents C. albicans mounting an appropriate oxidative stress response to amphotericin B, leading to fungal cell death. This work highlights the importance of understanding the impact of polymicrobial interactions on antimicrobial susceptibility.
Collapse
Affiliation(s)
- Farhana Alam
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Sarah A Blackburn
- Kent Fungal Group, Division of Natural Sciences, School of Biosciences, University of Kent, Canterbury, CT2 7NJ, UK
| | - Jack Davis
- Kent Fungal Group, Division of Natural Sciences, School of Biosciences, University of Kent, Canterbury, CT2 7NJ, UK
| | - Keely Massar
- Kent Fungal Group, Division of Natural Sciences, School of Biosciences, University of Kent, Canterbury, CT2 7NJ, UK
| | - Joao Correia
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Hung-Ji Tsai
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Jessica M A Blair
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Rebecca A Hall
- Kent Fungal Group, Division of Natural Sciences, School of Biosciences, University of Kent, Canterbury, CT2 7NJ, UK
| |
Collapse
|
13
|
MacAlpine J, Robbins N, Cowen LE. Bacterial-fungal interactions and their impact on microbial pathogenesis. Mol Ecol 2023; 32:2565-2581. [PMID: 35231147 PMCID: PMC11032213 DOI: 10.1111/mec.16411] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/14/2022] [Accepted: 02/18/2022] [Indexed: 11/27/2022]
Abstract
Microbial communities of the human microbiota exhibit diverse effects on human health and disease. Microbial homeostasis is important for normal physiological functions and changes to the microbiota are associated with many human diseases including diabetes, cancer, and colitis. In addition, there are many microorganisms that are either commensal or acquired from environmental reservoirs that can cause diverse pathologies. Importantly, the balance between health and disease is intricately connected to how members of the microbiota interact and affect one another's growth and pathogenicity. However, the mechanisms that govern these interactions are only beginning to be understood. In this review, we outline bacterial-fungal interactions in the human body, including examining the mechanisms by which bacteria govern fungal growth and virulence, as well as how fungi regulate bacterial pathogenesis. We summarize advances in the understanding of chemical, physical, and protein-based interactions, and their role in exacerbating or impeding human disease. We focus on the three fungal species responsible for the majority of systemic fungal infections in humans: Candida albicans, Cryptococcus neoformans, and Aspergillus fumigatus. We conclude by summarizing recent studies that have mined microbes for novel antimicrobials and antivirulence factors, highlighting the potential of the human microbiota as a rich resource for small molecule discovery.
Collapse
Affiliation(s)
- Jessie MacAlpine
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| |
Collapse
|
14
|
Traglia GM, Pasteran F, Escalante J, Nishimura B, Tuttobene MR, Subils T, Nuñez MR, Rivollier MG, Corso A, Tolmasky ME, Ramirez MS. Genomic Comparative Analysis of Two Multi-Drug Resistance (MDR) Acinetobacter baumannii Clinical Strains Assigned to International Clonal Lineage II Recovered Pre- and Post-COVID-19 Pandemic. BIOLOGY 2023; 12:358. [PMID: 36979049 PMCID: PMC10045941 DOI: 10.3390/biology12030358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/09/2023] [Accepted: 02/23/2023] [Indexed: 02/26/2023]
Abstract
BACKGROUND After the emergence of COVID-19, numerous cases of A. baumannii/SARS-CoV-2 co-infection were reported. Whether the co-infecting A. baumannii strains have distinctive characteristics remains unknown. METHODS AND RESULTS A. baumannii AMA_NO was isolated in 2021 from a patient with COVID-19. AMA166 was isolated from a mini-BAL used on a patient with pneumonia in 2016. Both genomes were similar, but they possessed 337 (AMA_NO) and 93 (AMA166) unique genes that were associated with biofilm formation, flagellar assembly, antibiotic resistance, secretion systems, and other functions. The antibiotic resistance genes were found within mobile genetic elements. While both strains harbored the carbapenemase-coding gene blaOXA-23, only the strain AMA_NO carried blaNDM-1. Representative functions coded for by virulence genes are the synthesis of the outer core of lipooligosaccharide (OCL5), biosynthesis and export of the capsular polysaccharide (KL2 cluster), high-efficiency iron uptake systems (acinetobactin and baumannoferrin), adherence, and quorum sensing. A comparative phylogenetic analysis including 239 additional sequence type (ST) 2 representative genomes showed high similarity to A. baumannii ABBL141. Since the degree of similarity that was observed between A. baumannii AMA_NO and AMA166 is higher than that found among other ST2 strains, we propose that they derive from a unique background based on core-genome phylogeny and comparative genome analysis. CONCLUSIONS Acquisition or shedding of specific genes could increase the ability of A. baumannii to infect patients with COVID-19.
Collapse
Affiliation(s)
- German Matias Traglia
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la Republica, Montevideo 11200, Uruguay
| | - Fernando Pasteran
- National Regional Reference Laboratory for Antimicrobial Resistance (NRL), Servicio Antimicrobianos, Instituto Nacional de Enfermedades Infecciosas, ANLIS Dr. Carlos G. Malbrán, Buenos Aires 1282, Argentina
| | - Jenny Escalante
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92831, USA
| | - Brent Nishimura
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92831, USA
| | - Marisel R. Tuttobene
- Instituto de Biología Molecular y Celular de Rosario (IBR, CONICET-UNR), Rosario 2000, Argentina
| | - Tomás Subils
- Instituto de Procesos Biotecnológicos y Químicos de Rosario (IPROBYQ, CONICET-UNR), Rosario 2000, Argentina
| | - Maria Rosa Nuñez
- Laboratorio de Microbiología, Hospital Provincial Neuquén Dr. Castro Rendón, Neuquén 8300, Argentina
| | | | - Alejandra Corso
- National Regional Reference Laboratory for Antimicrobial Resistance (NRL), Servicio Antimicrobianos, Instituto Nacional de Enfermedades Infecciosas, ANLIS Dr. Carlos G. Malbrán, Buenos Aires 1282, Argentina
| | - Marcelo E. Tolmasky
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92831, USA
| | - Maria Soledad Ramirez
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92831, USA
| |
Collapse
|
15
|
There Is More to Wounds than Bacteria: Fungal Biofilms in Chronic Wounds. CURRENT CLINICAL MICROBIOLOGY REPORTS 2023. [DOI: 10.1007/s40588-022-00187-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Abstract
Purpose of Review
The management of chronic wounds, a debilitating condition, presents a considerable challenge to healthcare professionals and a significant burden on services. When these wounds are exposed to the external environment, they are susceptible to microbial infection, which further complicates their management and worsens clinical outcomes.
Recent Findings
Bacteria typically exist in wounds as part of a biofilm, which is often polymicrobial in nature, alongside bacteria and fungi that are described as being more virulent and tolerant towards antimicrobials and antiseptics. Despite advancing knowledge in polymicrobial biofilm wound infections with respect to bacteria, the role of fungi is largely ignored, and their influence in chronicity and clinical management is not fully appreciated or understood.
Summary
The purpose of this review is to explore the significance of fungi within chronic wound environments and, in doing so, understand the importance of interkingdom interactions in wound management.
Collapse
|
16
|
Ahn SH, Karlsson AJ, Bentley WE, Raghavan SR. Capsules with bacteria and fungi in distinct compartments: A platform for studying microbes from different kingdoms and their cross-communication. PLoS One 2022; 17:e0277132. [DOI: 10.1371/journal.pone.0277132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 10/21/2022] [Indexed: 11/13/2022] Open
Abstract
Recently, we have created ‘artificial cells’ with an architecture mimicking that of typical eukaryotic cells. Our design uses common biopolymers like alginate and chitosan to create multi-compartment capsules (MCCs) via oil-free microfluidics. MCCs (~ 500 μm in diameter) can be engineered with multiple inner compartments, each with a distinct payload. This mimics the distinct organelles in eukaryotic cells, each of which has unique properties. In this study, we encapsulate microbial cells from two distinct kingdoms — Pseudomonas aeruginosa (bacteria) and Candida albicans (fungi) — in the inner compartments of MCCs. The two microbes are commonly found in biofilms at sites of infection in humans. We first demonstrate that the MCC can serve as a simple platform to observe the comparative growth of the cells in real time. Unlike typical co-culture in solution or on agar plates, the cells can grow in their own compartments without direct physical contact. Moreover, the hydrogel matrix in the compartments mimics the three-dimensional (3-D) environment that cells naturally encounter during their growth. Small molecules added to the solution are shown to permeate through the capsule walls and affect cell growth: for example, cationic surfactants inhibit the fungi but not the bacteria. Conversely, low pH and kanamycin inhibit the bacteria but not the fungi. Also, when the bacteria are present in adjacent compartments, the fungal cells mostly stay in a yeast morphology, meaning as spheroidal cells. In contrast, in the absence of the bacteria, the fungi transition into hyphae, i.e., long multicellular filaments. The inhibition of this morphological switch in fungal cells is shown to be induced by signaling molecules (specifically, the quorum sensing autoinducer-1 or AI-1) secreted by the bacteria. Thus, the MCC platform can also be used to detect cross-kingdom signaling between the compartmentalized microbes.
Collapse
|
17
|
Li H, Miao MX, Jia CL, Cao YB, Yan TH, Jiang YY, Yang F. Interactions between Candida albicans and the resident microbiota. Front Microbiol 2022; 13:930495. [PMID: 36204612 PMCID: PMC9531752 DOI: 10.3389/fmicb.2022.930495] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/31/2022] [Indexed: 01/09/2023] Open
Abstract
Candida albicans is a prevalent, opportunistic human fungal pathogen. It usually dwells in the human body as a commensal, however, once in its pathogenic state, it causes diseases ranging from debilitating superficial to life-threatening systemic infections. The switch from harmless colonizer to virulent pathogen is, in most cases, due to perturbation of the fungus-host-microbiota interplay. In this review, we focused on the interactions between C. albicans and the host microbiota in the mouth, gut, blood, and vagina. We also highlighted important future research directions. We expect that the evaluation of these interplays will help better our understanding of the etiology of fungal infections and shed new light on the therapeutic approaches.
Collapse
Affiliation(s)
- Hao Li
- Department of Pharmacy, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China,Department of Physiology and Pharmacology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ming-xing Miao
- Department of Physiology and Pharmacology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Cheng-lin Jia
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong-bing Cao
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tian-hua Yan
- Department of Physiology and Pharmacology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China,*Correspondence: Tian-hua Yan,
| | - Yuan-ying Jiang
- Department of Pharmacy, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China,Yuan-ying Jiang,
| | - Feng Yang
- Department of Pharmacy, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China,Feng Yang,
| |
Collapse
|
18
|
Phuengmaung P, Mekjaroen J, Saisorn W, Chatsuwan T, Somparn P, Leelahavanichkul A. Rapid Synergistic Biofilm Production of Pseudomonas and Candida on the Pulmonary Cell Surface and in Mice, a Possible Cause of Chronic Mixed Organismal Lung Lesions. Int J Mol Sci 2022; 23:ijms23169202. [PMID: 36012475 PMCID: PMC9409386 DOI: 10.3390/ijms23169202] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 11/16/2022] Open
Abstract
Due to the possible co-presence of Pseudomonas aeruginosa and Candida albicans (the most common nosocomial pathogens) in lungs, rapid interkingdom biofilm production is possible. As such, PA+CA produced more dominant biofilms on the pulmonary epithelial surface (NCI-H292) (confocal fluorescent extracellular matrix staining) with dominant psl upregulation, as demonstrated by polymerase chain reaction (PCR), after 8 h of experiments than PA alone. With a proteomic analysis, rhamnosyltransferase RhlB protein (Psl-associated quorum-sensing protein) was found to be among the high-abundance proteins in PA+CA than in PA biofilms, supporting psl-mediated biofilms in PA+CA on the cell surface. Additionally, PA+CA increased supernatant cytokines (IL-8 and IL-13, but not TNF-α, IL-6, and IL-10) with a similar upregulation of TLR-4, TLR-5, and TLR-9 (by PCR) compared with PA-stimulated cells. The intratracheal administration of PA+CA induced a greater severity of sepsis (serum creatinine, alanine transaminase, serum cytokines, and histology score) and prominent biofilms (fluorescent staining) with psl upregulation (PCR). In comparison with PA+CA biofilms on glass slides, PA+CA biofilms on biotic surfaces were more prominent (fluorescent staining). In conclusion, PA+CA induced Psl-predominant biofilms on the pulmonary cell surface and in mice with acute pneumonia, and these biofilms were more prominent than those induced by PA alone, highlighting the impact of Candida on rapid interkingdom biofilm production.
Collapse
Affiliation(s)
- Pornpimol Phuengmaung
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Jiradej Mekjaroen
- Center of Excellence in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Wilasinee Saisorn
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Tanittha Chatsuwan
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Poorichaya Somparn
- Center of Excellence in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: (P.S.); (A.L.); Tel.: +66-2-256-4251 (P.S. & A.L.); Fax: +66-2-252-6920 (P.S. & A.L.)
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: (P.S.); (A.L.); Tel.: +66-2-256-4251 (P.S. & A.L.); Fax: +66-2-252-6920 (P.S. & A.L.)
| |
Collapse
|
19
|
Liu L, Liu C, Cai J, Chen J, Chen J, Fu Y, Yi K, Wang H, Li X. Relationship Between Invasive Fungal Infection and Hypostatic Pneumonia: A Prospective Cohort Study. Front Microbiol 2022; 13:859359. [PMID: 35794923 PMCID: PMC9251507 DOI: 10.3389/fmicb.2022.859359] [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: 01/21/2022] [Accepted: 04/26/2022] [Indexed: 11/28/2022] Open
Abstract
Background The short-term mortality of hypostatic pneumonia (HP) is very high, and the treatment outcome is poor. The clinical diagnosis and treatment are primarily focused on bacterial and viral infection, ignoring the role of fungal infection at present. This study aims to validate the relationship between Invasive Fungal Infections (IFI) and HP. Methods In the cross-sectional study, a total of 11,371 participants have been enrolled. In the prospective cohort study, 4,441 individuals have been included at baseline and followed up from 2015 to 2019 with a total person years of 8,484.65. The standard procedures were used to assess questionnaire investigations, laboratory testing, and anthropometric indicators. For data analysis, logistic regression, restricted cubic spline, log-rank regression, Cox regression, and linear mixed effects model were applied to assess the relationship between IFI and HP risk longitudinally. Results In the cross-sectional study, elevated β-D-Glucan (BDG) concentrations are associated with a higher risk of HP prevalence in the total population, men, and women (ORT3 vs.T1 [95% CIs]: 2.12 [1.55, 2.91]; 2.01 [1.35, 2.99]; 2.34 [1.39, 3.94]), which were verified by a dose–effect relationship in the restricted cubic spline model. In the cohort study, Cox and Log-rank regression showed that the elevated BDG concentrations are associated with a significantly higher risk of HP incidence than participants with lower BDG concentrations (HRT3 vs.T1 [95% CIs]: 2.72 [1.36, 5.43], pLog–rank = 0.0086). During 5 years, the globulin (GLB) and C-reactive protein (CRP) were always on the top in the highest category of BDG concentrations. Between low and high BDG concentration, the total trend of GLB concentration was falling and the total trend of CRP concentration was rising with the increase of years (all the p-values < 0.0001). Conclusion In this study, IFI is associated with a higher risk of HP, with time sequence and related mechanisms requiring further investigation in the future.
Collapse
Affiliation(s)
- Lin Liu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chang Liu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianrong Cai
- Department of Ultrasound, Chongming Branch, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiayun Chen
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Chen
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanyuan Fu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kexin Yi
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Wang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Hui Wang,
| | - Xue Li
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Xue Li,
| |
Collapse
|
20
|
Pohl CH. Recent Advances and Opportunities in the Study of Candida albicans Polymicrobial Biofilms. Front Cell Infect Microbiol 2022; 12:836379. [PMID: 35252039 PMCID: PMC8894716 DOI: 10.3389/fcimb.2022.836379] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/26/2022] [Indexed: 01/11/2023] Open
Abstract
It is well known that the opportunistic pathogenic yeast, Candida albicans, can form polymicrobial biofilms with a variety of bacteria, both in vitro and in vivo, and that these polymicrobial biofilms can impact the course and management of disease. Although specific interactions are often described as either synergistic or antagonistic, this may be an oversimplification. Polymicrobial biofilms are complex two-way interacting communities, regulated by inter-domain (inter-kingdom) signaling and various molecular mechanisms. This review article will highlight advances over the last six years (2016-2021) regarding the unique biology of polymicrobial biofilms formed by C. albicans and bacteria, including regulation of their formation. In addition, some of the consequences of these interactions, such as the influence of co-existence on antimicrobial susceptibility and virulence, will be discussed. Since the aim of this knowledge is to inform possible alternative treatment options, recent studies on the discovery of novel anti-biofilm compounds will also be included. Throughout, an attempt will be made to identify ongoing challenges in this area.
Collapse
|
21
|
Ahmed A, Getti G, Boateng J. Medicated multi-targeted alginate-based dressings for potential treatment of mixed bacterial-fungal infections in diabetic foot ulcers. Int J Pharm 2021; 606:120903. [PMID: 34293470 DOI: 10.1016/j.ijpharm.2021.120903] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 01/13/2023]
Abstract
Recently developed medicated dressings target either bacterial or fungal infection only, which is not effective for the treatment of mixed infections common in diabetic foot ulcers (DFUs). This study aimed to develop advanced bioactive alginate-based dressings (films and wafers) to deliver therapeutically relevant doses of ciprofloxacin (CIP) and fluconazole (FLU) to target mixed bacterial and fungal infections in DFUs. The alginate compatibility with the drugs was confirmed by SEM, XRD, FTIR and texture analysis, while the medicated wafers showed better fluid handling properties than the films in the presence of simulated wound fluid. The dressings showed initial fast release of FLU followed by sustained release of CIP which completely eradicated E. coli, S. aureus, P. aeruginosa and reduced fungal load (C. albicans) by 10-fold within 24 h. Moreover, the medicated dressings were biocompatible (>70% cell viability over 72 h) with human primary adult keratinocytes and in-vitro scratch assay showed 65-68% wound closure within 7 days.
Collapse
Affiliation(s)
- Asif Ahmed
- School of Science, Faculty of Engineering and Science, University of Greenwich, Medway, Central Ave., Chatham Maritime, Kent ME4 4TB, UK
| | - Giulia Getti
- School of Science, Faculty of Engineering and Science, University of Greenwich, Medway, Central Ave., Chatham Maritime, Kent ME4 4TB, UK
| | - Joshua Boateng
- School of Science, Faculty of Engineering and Science, University of Greenwich, Medway, Central Ave., Chatham Maritime, Kent ME4 4TB, UK.
| |
Collapse
|
22
|
Light Modulates Important Pathogenic Determinants and Virulence in ESKAPE Pathogens Acinetobacter baumannii, Pseudomonas aeruginosa, and Staphylococcus aureus. J Bacteriol 2021; 203:JB.00566-20. [PMID: 33288627 DOI: 10.1128/jb.00566-20] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023] Open
Abstract
Light sensing has been extensively characterized in the human pathogen Acinetobacter baumannii at environmental temperatures. However, the influence of light on the physiology and pathogenicity of human bacterial pathogens at temperatures found in warm-blooded hosts is still poorly understand. In this work, we show that Staphylococcus aureus, Acinetobacter baumannii, and Pseudomonas aeruginosa (ESKAPE) priority pathogens, which have been recognized by the WHO and the CDC as critical, can also sense and respond to light at temperatures found in human hosts. Most interestingly, in these pathogens, light modulates important pathogenicity determinants as well as virulence in an epithelial infection model, which could have implications in human infections. In fact, we found that alpha-toxin-dependent hemolysis, motility, and growth under iron-deprived conditions are modulated by light in S. aureus Light also regulates persistence, metabolism, and the ability to kill competitors in some of these microorganisms. Finally, light exerts a profound effect on the virulence of these pathogens in an epithelial infection model, although the response is not the same in the different species; virulence was enhanced by light in A. baumannii and S. aureus, while in A. nosocomialis and P. aeruginosa it was reduced. Neither the BlsA photoreceptor nor the type VI secretion system (T6SS) is involved in virulence modulation by light in A. baumannii Overall, this fundamental knowledge highlights the potential use of light to control pathogen virulence, either directly or by manipulating the light regulatory switch toward the lowest virulence/persistence configuration.IMPORTANCE Pathogenic bacteria are microorganisms capable of producing disease. Dangerous bacterial pathogens, such as Staphylococcus aureus, Pseudomonas aeruginosa, and Acinetobacter baumannii, are responsible for serious intrahospital and community infections in humans. Therapeutics is often complicated due to resistance to multiple antibiotics, rendering them ineffective. In this work, we show that these pathogens sense natural light and respond to it by modulating aspects related to their ability to cause disease; in the presence of light, some of them become more aggressive, while others show an opposite response. Overall, we provide new understanding on the behavior of these pathogens, which could contribute to the control of infections caused by them. Since the response is distributed in diverse pathogens, this notion could prove a general concept.
Collapse
|
23
|
Gaston JR, Johnson AO, Bair KL, White AN, Armbruster CE. Polymicrobial interactions in the urinary tract: is the enemy of my enemy my friend? Infect Immun 2021; 89:IAI.00652-20. [PMID: 33431702 DOI: 10.1128/iai.00652-20] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The vast majority of research pertaining to urinary tract infection has focused on a single pathogen in isolation, and predominantly Escherichia coli. However, polymicrobial urine colonization and infection are prevalent in several patient populations, including individuals with urinary catheters. The progression from asymptomatic colonization to symptomatic infection and severe disease is likely shaped by interactions between traditional pathogens as well as constituents of the normal urinary microbiota. Recent studies have begun to experimentally dissect the contribution of polymicrobial interactions to disease outcomes in the urinary tract, including their role in development of antimicrobial-resistant biofilm communities, modulating the innate immune response, tissue damage, and sepsis. This review aims to summarize the epidemiology of polymicrobial urine colonization, provide an overview of common urinary tract pathogens, and present key microbe-microbe and host-microbe interactions that influence infection progression, persistence, and severity.
Collapse
Affiliation(s)
- Jordan R Gaston
- Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo
| | - Alexandra O Johnson
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo
| | - Kirsten L Bair
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo
| | - Ashley N White
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo
| | - Chelsie E Armbruster
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo
| |
Collapse
|
24
|
Abd El-Baky RM, Mandour SA, Ahmed EF, Hashem ZS, Sandle T, Mohamed DS. Virulence profiles of some Pseudomonas aeruginosa clinical isolates and their association with the suppression of Candida growth in polymicrobial infections. PLoS One 2020; 15:e0243418. [PMID: 33290412 PMCID: PMC7723275 DOI: 10.1371/journal.pone.0243418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 11/22/2020] [Indexed: 11/18/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that can cause a variety of diseases especially in the hospital environment. However, this pathogen also exhibits antimicrobial activity against Gram-positive bacteria and fungi. This study aimed to characterize different virulence factors, secreted metabolites and to study their role in the suppression of Candida growth. Fifteen P. aeruginosa isolates were tested for their anticandidal activity against 3 different Candida spp. by the cross-streak method. The effect on hyphae production was tested microscopically using light and scanning electron microscopy (SEM). Polymerase chain reaction was used in the detection of some virulence genes. Lipopolysaccharide profile was performed using SDS-polyacrylamide gel stained with silver. Fatty acids were analyzed by GC-MS as methyl ester derivatives. It was found that 5 P. aeruginosa isolates inhibited all tested Candida spp. (50-100% inhibition), one isolate inhibited C. glabrata only and 3 isolates showed no activity against the tested Candida spp. The P. aeruginosa isolates inhibiting all Candida spp. were positive for all virulence genes. GC-Ms analysis revealed that isolates with high anticandidal activity showed spectra for several compounds, each known for their antifungal activity in comparison to those with low or no anticandidal activity. Hence, clinical isolates of P. aeruginosa showed Candida species-specific interactions by different means, giving rise to the importance of studying microbial interaction in polymicrobial infections and their contribution to causing disease.
Collapse
Affiliation(s)
- Rehab Mahmoud Abd El-Baky
- Department of Microbiology and Immunology, Faculty of Pharmacy, Minia University, Minia, Egypt
- Department of Microbiology and Immunology, Faculty of Pharmacy, Deraya University, Minia, Egypt
| | - Sahar A. Mandour
- Department of Microbiology and Immunology, Faculty of Pharmacy, Deraya University, Minia, Egypt
| | - Eman Farouk Ahmed
- Department of Microbiology and Immunology, Faculty of Pharmacy, Deraya University, Minia, Egypt
| | - Zeinab Shawky Hashem
- Department of Microbiology and Immunology, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Tim Sandle
- School of Health Sciences, Division of Pharmacy & Optometry, University of Manchester, Manchester, United Kingdom
| | - Doaa Safwat Mohamed
- Department of Microbiology and Immunology, Faculty of Pharmacy, Deraya University, Minia, Egypt
| |
Collapse
|
25
|
Phuengmaung P, Somparn P, Panpetch W, Singkham-In U, Wannigama DL, Chatsuwan T, Leelahavanichkul A. Coexistence of Pseudomonas aeruginosa With Candida albicans Enhances Biofilm Thickness Through Alginate-Related Extracellular Matrix but Is Attenuated by N-acetyl-l-cysteine. Front Cell Infect Microbiol 2020; 10:594336. [PMID: 33330136 PMCID: PMC7732535 DOI: 10.3389/fcimb.2020.594336] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/15/2020] [Indexed: 01/25/2023] Open
Abstract
Bacteria and Candidaalbicans are prominent gut microbiota, and the translocation of these organisms into blood circulation might induce mixed-organism biofilms, which warrants the exploration of mixed- versus single-organism biofilms in vitro and in vivo. In single-organism biofilms, Acinetobacter baumannii and Pseudomonas aeruginosa (PA) produced the least and the most prominent biofilms, respectively. C. albicans with P. aeruginosa (PA+CA) induced the highest biofilms among mixed-organism groups as determined by crystal violet straining. The sessile form of PA+CA induced higher macrophage responses than sessile PA, which supports enhanced immune activation toward mixed-organism biofilms. In addition, Candida incubated in pre-formed Pseudomonas biofilms (PA>CA) produced even higher biofilms than PA+CA (simultaneous incubation of both organisms) as determined by fluorescent staining on biofilm matrix (AF647 color). Despite the initially lower bacteria during preparation, bacterial burdens by culture in mixed-organism biofilms (PA+CA and PA>CA) were not different from biofilms of PA alone, supporting Candida-enhanced Pseudomonas growth. Moreover, proteomic analysis in PA>CA biofilms demonstrated high AlgU and mucA with low mucB when compared with PA alone or PA+CA, implying an alginate-related mucoid phenotype in PA>CA biofilms. Furthermore, mice with PA>CA biofilms demonstrated higher bacteremia with more severe sepsis compared with mice with PA+CA biofilms. This is possibly due to the different structures. Interestingly, l-cysteine, a biofilm matrix inhibitor, attenuated mixed-organism biofilms both in vitro and in mice. In conclusion, Candida enhanced Pseudomonas alginate–related biofilm production, and Candida presentation in pre-formed Pseudomonas biofilms might alter biofilm structures that affect clinical manifestations but was attenuated by l-cysteine.
Collapse
Affiliation(s)
- Pornpimol Phuengmaung
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Poorichaya Somparn
- Center of Excellence in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Wimonrat Panpetch
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Uthaibhorn Singkham-In
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Tanittha Chatsuwan
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
26
|
Grainha T, Jorge P, Alves D, Lopes SP, Pereira MO. Unraveling Pseudomonas aeruginosa and Candida albicans Communication in Coinfection Scenarios: Insights Through Network Analysis. Front Cell Infect Microbiol 2020; 10:550505. [PMID: 33262953 PMCID: PMC7686562 DOI: 10.3389/fcimb.2020.550505] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 10/12/2020] [Indexed: 12/19/2022] Open
Abstract
Modern medicine is currently facing huge setbacks concerning infection therapeutics as microorganisms are consistently knocking down every antimicrobial wall set before them. The situation becomes more worrying when taking into account that, in both environmental and disease scenarios, microorganisms present themselves as biofilm communities that are often polymicrobial. This comprises a competitive advantage, with interactions between different species altering host responses, antimicrobial effectiveness, microbial pathogenesis and virulence, usually augmenting the severity of the infection and contributing for the recalcitrance towards conventional therapy. Pseudomonas aeruginosa and Candida albicans are two opportunistic pathogens often co-isolated from infections, mainly from mucosal tissues like the lung. Despite the billions of years of co-existence, this pair of microorganisms is a great example on how little is known about cross-kingdom interactions, particularly within the context of coinfections. Given the described scenario, this study aimed to collect, curate, and analyze all published experimental information on the molecular basis of P. aeruginosa and C. albicans interactions in biofilms, in order to shed light into key mechanisms that may affect infection prognosis, increasing this area of knowledge. Publications were optimally retrieved from PubMed and Web of Science and classified as to their relevance. Data was then systematically and manually curated, analyzed, and further reconstructed as networks. A total of 641 interactions between the two pathogens were annotated, outputting knowledge on important molecular players affecting key virulence mechanisms, such as hyphal growth, and related genes and proteins, constituting potential therapeutic targets for infections related to these bacterial-fungal consortia. Contrasting interactions were also analyzed, and quorum-sensing inhibition approaches were highlighted. All annotated data was made publicly available at www.ceb.uminho.pt/ISCTD, a database already containing similar data for P. aeruginosa and Staphylococcus aureus communication. This will allow researchers to cut on time and effort when studying this particular subject, facilitating the understanding of the basis of the inter-species and inter-kingdom interactions and how it can be modulated to help design alternative and more effective tailored therapies. Finally, data deposition will serve as base for future dataset integration, whose analysis will hopefully give insights into communications in more complex and varied biofilm communities.
Collapse
Affiliation(s)
- Tânia Grainha
- CEB-Centre of Biological Engineering, LIBRO-Laboratory of Research in Biofilms Rosário Oliveira, University of Minho, Campus de Gualtar, Braga, Portugal
| | - Paula Jorge
- CEB-Centre of Biological Engineering, LIBRO-Laboratory of Research in Biofilms Rosário Oliveira, University of Minho, Campus de Gualtar, Braga, Portugal
| | - Diana Alves
- CEB-Centre of Biological Engineering, LIBRO-Laboratory of Research in Biofilms Rosário Oliveira, University of Minho, Campus de Gualtar, Braga, Portugal
| | - Susana Patrícia Lopes
- CEB-Centre of Biological Engineering, LIBRO-Laboratory of Research in Biofilms Rosário Oliveira, University of Minho, Campus de Gualtar, Braga, Portugal
| | - Maria Olívia Pereira
- CEB-Centre of Biological Engineering, LIBRO-Laboratory of Research in Biofilms Rosário Oliveira, University of Minho, Campus de Gualtar, Braga, Portugal
| |
Collapse
|
27
|
Bandara HMHN, Wood DLA, Vanwonterghem I, Hugenholtz P, Cheung BPK, Samaranayake LP. Fluconazole resistance in Candida albicans is induced by Pseudomonas aeruginosa quorum sensing. Sci Rep 2020; 10:7769. [PMID: 32385378 PMCID: PMC7211000 DOI: 10.1038/s41598-020-64761-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 04/22/2020] [Indexed: 12/16/2022] Open
Abstract
Microorganisms employ quorum sensing (QS) mechanisms to communicate with each other within microbial ecosystems. Emerging evidence suggests that intraspecies and interspecies QS plays an important role in antimicrobial resistance in microbial communities. However, the relationship between interkingdom QS and antimicrobial resistance is largely unknown. Here, we demonstrate that interkingdom QS interactions between a bacterium, Pseudomonas aeruginosa and a yeast, Candida albicans, induce the resistance of the latter to a widely used antifungal fluconazole. Phenotypic, transcriptomic, and proteomic analyses reveal that P. aeruginosa's main QS molecule, N-(3-Oxododecanoyl)-L-homoserine lactone, induces candidal resistance to fluconazole by reversing the antifungal's effect on the ergosterol biosynthesis pathway. Accessory resistance mechanisms including upregulation of C. albicans drug-efflux, regulation of oxidative stress response, and maintenance of cell membrane integrity, further confirm this phenomenon. These findings demonstrate that P. aeruginosa QS molecules may confer protection to neighboring yeasts against azoles, in turn strengthening their co-existence in hostile polymicrobial infection sites.
Collapse
Affiliation(s)
- H M H N Bandara
- Oral Microbiology, Bristol Dental School, University of Bristol, Lower Maudlin Street, Bristol, BS1 2LY, UK.
| | - D L A Wood
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | - I Vanwonterghem
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | - P Hugenholtz
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | - B P K Cheung
- Faculty of Dentistry, The University of Hong Kong, 34 Hospital Rd, Sai Ying Pun, Hong Kong SAR, China
| | - L P Samaranayake
- College of Dental Medicine, The University of Sharjah, P.O. Box, 27272, Sharjah, UAE
| |
Collapse
|
28
|
Alam F, Catlow D, Di Maio A, Blair JMA, Hall RA. Candida albicans enhances meropenem tolerance of Pseudomonas aeruginosa in a dual-species biofilm. J Antimicrob Chemother 2020; 75:925-935. [PMID: 31865379 PMCID: PMC7069478 DOI: 10.1093/jac/dkz514] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 10/18/2019] [Accepted: 11/12/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Pseudomonas aeruginosa is an opportunistic bacterium that infects the airways of cystic fibrosis patients, surfaces of surgical and burn wounds, and indwelling medical devices. Patients are prone to secondary fungal infections, with Candida albicans being commonly co-isolated with P. aeruginosa. Both P. aeruginosa and C. albicans are able to form extensive biofilms on the surfaces of mucosa and medical devices. OBJECTIVES To determine whether the presence of C. albicans enhances antibiotic tolerance of P. aeruginosa in a dual-species biofilm. METHODS Single- and dual-species biofilms were established in microtitre plates and the survival of each species was measured following treatment with clinically relevant antibiotics. Scanning electron microscopy and confocal microscopy were used to visualize biofilm structure. RESULTS C. albicans enhances P. aeruginosa biofilm tolerance to meropenem at the clinically relevant concentration of 5 mg/L. This effect is specific to biofilm cultures and is dependent upon C. albicans extracellular matrix polysaccharides, mannan and glucan, with C. albicans cells deficient in glycosylation structures not enhancing P. aeruginosa tolerance to meropenem. CONCLUSIONS We propose that fungal mannan and glucan secreted into the extracellular matrix of P. aeruginosa/C. albicans dual-species biofilms play a central role in enhancing P. aeruginosa tolerance to meropenem, which has direct implications for the treatment of coinfected patients.
Collapse
Affiliation(s)
- Farhana Alam
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Dominic Catlow
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Alessandro Di Maio
- Birmingham Advanced Light Microscopy, School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Jessica M A Blair
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Rebecca A Hall
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
- Kent Fungal Group, School of Biosciences, University of Kent, Canterbury, CT2 7NZ, UK
| |
Collapse
|
29
|
Azzam SZ, Cayme GJ, Martinez LR. Polymicrobial interactions involving fungi and their importance for the environment and in human disease. Microb Pathog 2020; 140:103942. [PMID: 31881258 PMCID: PMC7105396 DOI: 10.1016/j.micpath.2019.103942] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/17/2019] [Accepted: 12/19/2019] [Indexed: 01/08/2023]
Abstract
Understanding polymicrobial interactions involving fungi in the environment and the human mycobiome is necessary to address environmental and medically related problems such as drought or antimicrobial resistance. The diversity of these interactions highlights the complexity of fungi, considering how some interactions can be antagonistic, while others synergistic. Over the years, an increase in studies on the mycobiome have revealed similarities between the human and environmental hosts. More recently, studies have focused on microbial commensal relationships and identifying causative agents of human disease. The overlap of some of these interactions is impossible to ignore, indicating that there are areas for medical exploitation that need to be further investigated. This review provides the latest advances in polymicrobial interactions involving fungi and discusses the importance of the fungal lifestyle in the environment and in human disease.
Collapse
Affiliation(s)
- Seham Z Azzam
- Department of Biological Sciences, The Border Biomedical Research Center, University of Texas at El Paso, TX, USA
| | - Ginelle J Cayme
- Department of Biological Sciences, The Border Biomedical Research Center, University of Texas at El Paso, TX, USA
| | - Luis R Martinez
- Department of Biological Sciences, The Border Biomedical Research Center, University of Texas at El Paso, TX, USA; Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
30
|
She P, Liu Y, Wang Y, Tan F, Luo Z, Wu Y. Antibiofilm efficacy of the gold compound auranofin on dual species biofilms of
Staphylococcus aureus
and
Candida
sp. J Appl Microbiol 2019; 128:88-101. [PMID: 31509623 DOI: 10.1111/jam.14443] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/12/2019] [Accepted: 08/31/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Pengfei She
- Department of Clinical Laboratory The Third Xiangya Hospital of Central South University Changsha P.R. China
| | - Yiqing Liu
- Department of Clinical Laboratory The Third Xiangya Hospital of Central South University Changsha P.R. China
| | - Yangxia Wang
- Department of Clinical Laboratory The First Affiliated Hospital of Zhengzhou University Zhengzhou P.R. China
| | - Fang Tan
- Department of Clinical Laboratory The Third Xiangya Hospital of Central South University Changsha P.R. China
| | - Zhen Luo
- Department of Clinical Laboratory The Third Xiangya Hospital of Central South University Changsha P.R. China
| | - Yong Wu
- Department of Clinical Laboratory The Third Xiangya Hospital of Central South University Changsha P.R. China
| |
Collapse
|
31
|
Interactions between Aspergillus fumigatus and Pulmonary Bacteria: Current State of the Field, New Data, and Future Perspective. J Fungi (Basel) 2019; 5:jof5020048. [PMID: 31212791 PMCID: PMC6617096 DOI: 10.3390/jof5020048] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/10/2019] [Accepted: 06/10/2019] [Indexed: 12/14/2022] Open
Abstract
Aspergillus fumigatus and Pseudomonas aeruginosa are central fungal and bacterial members of the pulmonary microbiota. The interactions between A. fumigatus and P. aeruginosa have only just begun to be explored. A balance between inhibitory and stimulatory effects on fungal growth was observed in mixed A. fumigatus-P. aeruginosa cultures. Negative interactions have been seen for homoserine-lactones, pyoverdine and pyochelin resulting from iron starvation and intracellular inhibitory reactive oxidant production. In contrast, several types of positive interactions were recognized. Dirhamnolipids resulted in the production of a thick fungal cell wall, allowing the fungus to resist stress. Phenazines and pyochelin favor iron uptake for the fungus. A. fumigatus is able to use bacterial volatiles to promote its growth. The immune response is also differentially regulated by co-infections.
Collapse
|
32
|
Low concentrations of acetic and formic acids enhance the inactivation of Staphylococcus aureus and Pseudomonas aeruginosa with pulsed electric fields. BMC Microbiol 2019; 19:73. [PMID: 30943901 PMCID: PMC6448289 DOI: 10.1186/s12866-019-1447-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 03/28/2019] [Indexed: 02/07/2023] Open
Abstract
Background Skin infections, particularly caused by drug-resistant pathogens, represent a clinical challenge due to being a frequent cause of morbidity and mortality. The objectives of this study were to examine if low concentrations of acetic and formic acids can increase sensitivity of Staphylococcus aureus and Pseudomonas aeruginosa to pulsed electric field (PEF) and thus, promote a fast and efficient treatment methodology for wound treatment. Results We have shown that the combination of PEF (10–30 kV/cm) with organic acids (0.1% formic and acetic acids) increased the bactericidal properties of treatment. The effect was apparent for both acids. The proposed methodology allowed to reduce the energy of electrical pulses and the inhibitory concentrations of acids, while still maintain high efficiency of bacteria eradication. Conclusions Application of weak organic acids as bactericidal agents has many advantages over antibiotics because they do not trigger development of drug-resistance in bacteria. The combination with PEF can make the treatment effective even against biofilms. The results of this study are particularly useful for the development of new methodologies for the treatment of extreme cases of wound infections when the chemical treatment is no longer effective or hinders wound healing.
Collapse
|
33
|
Deveau A, Bonito G, Uehling J, Paoletti M, Becker M, Bindschedler S, Hacquard S, Hervé V, Labbé J, Lastovetsky OA, Mieszkin S, Millet LJ, Vajna B, Junier P, Bonfante P, Krom BP, Olsson S, van Elsas JD, Wick LY. Bacterial-fungal interactions: ecology, mechanisms and challenges. FEMS Microbiol Rev 2018; 42:335-352. [PMID: 29471481 DOI: 10.1093/femsre/fuy008] [Citation(s) in RCA: 381] [Impact Index Per Article: 54.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 02/16/2018] [Indexed: 12/14/2022] Open
Abstract
Fungi and bacteria are found living together in a wide variety of environments. Their interactions are significant drivers of many ecosystem functions and are important for the health of plants and animals. A large number of fungal and bacterial families engage in complex interactions that lead to critical behavioural shifts of the microorganisms ranging from mutualism to antagonism. The importance of bacterial-fungal interactions (BFI) in environmental science, medicine and biotechnology has led to the emergence of a dynamic and multidisciplinary research field that combines highly diverse approaches including molecular biology, genomics, geochemistry, chemical and microbial ecology, biophysics and ecological modelling. In this review, we discuss recent advances that underscore the roles of BFI across relevant habitats and ecosystems. A particular focus is placed on the understanding of BFI within complex microbial communities and in regard of the metaorganism concept. We also discuss recent discoveries that clarify the (molecular) mechanisms involved in bacterial-fungal relationships, and the contribution of new technologies to decipher generic principles of BFI in terms of physical associations and molecular dialogues. Finally, we discuss future directions for research in order to stimulate synergy within the BFI research area and to resolve outstanding questions.
Collapse
Affiliation(s)
- Aurélie Deveau
- Université de Lorraine, INRA, UMR IAM, 54280 Champenoux, France
| | - Gregory Bonito
- Department of Plant Soil and Microbial Sciences, Michigan State University, East Lansing, MI 48824, USA
| | - Jessie Uehling
- Biology Department, Duke University, Box 90338, Durham, NC 27705, USA.,Plant and Microbial Biology, University of California, Berkeley, CA 94703, USA
| | - Mathieu Paoletti
- Institut de Biologie et Génétique Cellulaire, UMR 5095 CNRS et Université de Bordeaux, 1 rue Camille Saint-Saëns, 33077 Bordeaux cedex, France
| | - Matthias Becker
- IGZ, Leibniz-Institute of Vegetable and Ornamental Crops, 14979 Großbeeren, Germany
| | - Saskia Bindschedler
- Laboratory of Microbiology, Institute of Biology, University of Neuchâtel, CH-2000 Neuchâtel, Switzerland
| | - Stéphane Hacquard
- Department of Plant Microbe Interactions, Max Planck Institute for Plant Breeding Research, 50829 Cologne, Germany
| | - Vincent Hervé
- Laboratory of Microbiology, Institute of Biology, University of Neuchâtel, CH-2000 Neuchâtel, Switzerland.,Laboratory of Biogeosciences, Institute of Earth Surface Dynamics, University of Lausanne, CH-1015 Lausanne, Switzerland
| | - Jessy Labbé
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN 37831, USA.,Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - Olga A Lastovetsky
- Graduate Field of Microbiology, Cornell University, Ithaca, NY 14853, USA
| | - Sophie Mieszkin
- Université de Lorraine, INRA, UMR IAM, 54280 Champenoux, France
| | - Larry J Millet
- Joint Institute for Biological Science, University of Tennessee, and the Biosciences Division of Oak Ridge National Laboratory, Oak Ridge, TN 37831, USA
| | - Balázs Vajna
- Department of Microbiology, Eötvös Loránd University, Pázmány Péter sétány 1/C, 1117 Budapest, Hungary
| | - Pilar Junier
- Laboratory of Microbiology, Institute of Biology, University of Neuchâtel, CH-2000 Neuchâtel, Switzerland
| | - Paola Bonfante
- Department of Life Science and Systems Biology, University of Torino, 10125 Torino, Italy
| | - Bastiaan P Krom
- Department of Preventive Dentistry, Academic Centre for Dentistry, G. Mahlerlaan 3004, 1081 LA, Amsterdam, The Netherlands
| | - Stefan Olsson
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University (FAFU), Fuzhou 350002, China
| | - Jan Dirk van Elsas
- Microbial Ecology group, GELIFES, University of Groningen, 9747 Groningen, The Netherlands
| | - Lukas Y Wick
- Helmholtz Centre for Environmental Research-UFZ, Department of Environmental Microbiology, Permoserstraße 15, 04318 Leipzig, Germany
| |
Collapse
|
34
|
Padder SA, Prasad R, Shah AH. Quorum sensing: A less known mode of communication among fungi. Microbiol Res 2018; 210:51-58. [PMID: 29625658 DOI: 10.1016/j.micres.2018.03.007] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 02/21/2018] [Accepted: 03/17/2018] [Indexed: 02/08/2023]
Abstract
Quorum sensing (QS), a density-dependent signaling mechanism of microbial cells, involves an exchange and sense of low molecular weight signaling compounds called autoinducers. With the increase in population density, the autoinducers accumulate in the extracellular environment and once their concentration reaches a threshold, many genes are either expressed or repressed. This cell density-dependent signaling mechanism enables single cells to behave as multicellular organisms and regulates different microbial behaviors like morphogenesis, pathogenesis, competence, biofilm formation, bioluminescence, etc guided by environmental cues. Initially, QS was regarded to be a specialized system of certain bacteria. The discovery of filamentation control in pathogenic polymorphic fungus Candida albicans by farnesol revealed the phenomenon of QS in fungi as well. Pathogenic microorganisms primarily regulate the expression of virulence genes using QS systems. The indirect role of QS in the emergence of multiple drug resistance (MDR) in microbial pathogens necessitates the finding of alternative antimicrobial therapies that target QS and inhibit the same. A related phenomenon of quorum sensing inhibition (QSI) performed by small inhibitor molecules called quorum sensing inhibitors (QSIs) has an ability for efficient reduction of gene expression regulated by quorum sensing. In the present review, recent advancements in the study of different fungal quorum sensing molecules (QSMs) and quorum sensing inhibitors (QSIs) of fungal origin along with their mechanism of action and/or role/s are discussed.
Collapse
Affiliation(s)
- Sajad Ahmad Padder
- Department of Bioresources, University of Kashmir, Hazratbal, Srinagar 190006, J&K, India
| | - Rajendra Prasad
- Amity Institute of Integrative Sciences and Health and Amity Institute of Biotechnology, Amity University Haryana, Amity Education Valley, Gurgaon 122413, HR, India
| | - Abdul Haseeb Shah
- Department of Bioresources, University of Kashmir, Hazratbal, Srinagar 190006, J&K, India.
| |
Collapse
|