Microbial Interactions during Infection

A special issue of Pathogens (ISSN 2076-0817).

Deadline for manuscript submissions: closed (31 December 2020) | Viewed by 60180

Special Issue Editors


E-Mail Website
Guest Editor
Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA
Interests: biofilm physiology; polymicrobial interactions; antibiotic drug discovery; microbial evolution; host/pathogen interactions

E-Mail Website
Co-Guest Editor
Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida 32610, United States
Interests: Biofilms, Environment, Fungi, Infections, Polymicrobial interactions

Special Issue Information

Dear Colleagues,

As Guest Editor of a Special Issue of Pathogens focused on microbial interactions during infection, I invite you, as an expert in this field, to submit a manuscript for publication.

In this issue, we seek to highlight both research articles as well as review articles elucidating the importance of polymicrobial interactions in shaping disease progression and therapeutic efficacy. The types of interactions of interest range from the synergistic actions of multiple pathogens leading to increased disease severity and/or antimicrobial recalcitrance to antagonistic actions of commensals protecting the host from invading pathogens. In general, the purpose of this issue is to highlight the diverse types of polymicrobial interactions that can occur during infection, the impact of these interactions on microbe or host physiology, and how these interactions should be taken into consideration when studying infectious diseases. Until recently, the bulk of literature in the area of disease progression and therapeutic discovery was focused on single species infection models despite the evidence that a large number of human infections contain a diverse mixture of microorganisms, including viruses, bacteria, and fungi. In the past few years, there have been tremendous contributions to the study of polymicrobial infections that have highlighted the importance of this clinically-relevant phenomenon. Therefore, we believe that this type of modern focus in the area of infectious disease warrants a Special Issue of Pathogens.

Dr. Catherine Wakeman
Dr. Luis R. Martinez
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Pathogens is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • host–microbe interactions
  • polymicrobial interactions
  • biofilm
  • immune response
  • antibiotic discovery

Published Papers (16 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review, Other

12 pages, 4561 KiB  
Article
Intracellular Microbiome Profiling of the Acanthamoeba Clinical Isolates from Lens Associated Keratitis
by Yu-Jen Wang, Sung-Chou Li, Wei-Chen Lin and Fu-Chin Huang
Pathogens 2021, 10(3), 266; https://doi.org/10.3390/pathogens10030266 - 25 Feb 2021
Cited by 9 | Viewed by 1809
Abstract
Acanthamoeba act as hosts for various microorganisms and pathogens, causing Acanthamoeba Keratitis (AK). To investigate the association between endosymbionts and AK progression, we performed a metagenomics study to characterize the intracellular microbiome from five lenses associated with AK isolates and standard strains to [...] Read more.
Acanthamoeba act as hosts for various microorganisms and pathogens, causing Acanthamoeba Keratitis (AK). To investigate the association between endosymbionts and AK progression, we performed a metagenomics study to characterize the intracellular microbiome from five lenses associated with AK isolates and standard strains to characterize the role of ocular flora in AK progression. The used clinical isolates were axenic cultured from lenses associated with AK patients. AK isolates and standard controls such as 16S ribosomal RNA sequencing techniques were used for analysis. The microbiome compositions and relative abundance values were compared. The orders of Clostridiales and Bacteroidales presented major populations of intracellular microbes belonging to all isolates. Comparison of the different source isolates showed that most of the abundance in keratitis isolates came from Ruminococcus gnavus (121.0 folds), Eubacterium dolichum (54.15 folds), Roseburia faecis (24.51 folds), and Blautia producta (3.15 folds). Further analysis of the relative abundance data from keratitis isolates showed that Blautia producta was positively correlated with the disease course. In contrast, Bacteroides ovatus was found to be abundant in early-stage keratitis isolates. This study reveals the abundant anaerobic Gram-positive rods present in severe keratitis isolate and characterize the association between Acanthamoeba and ocular flora in AK progression. Full article
(This article belongs to the Special Issue Microbial Interactions during Infection)
Show Figures

Figure 1

15 pages, 4640 KiB  
Article
Acinetobacter baumannii Strains Deficient in the Clp Chaperone-Protease Genes Have Reduced Virulence in a Murine Model of Pneumonia
by J Christian Belisario, Hiu Ham Lee, Harshani Luknauth, Nathan W. Rigel and Luis R. Martinez
Pathogens 2021, 10(2), 204; https://doi.org/10.3390/pathogens10020204 - 13 Feb 2021
Cited by 3 | Viewed by 3433
Abstract
Acinetobacter baumannii has emerged as a significant opportunistic Gram-negative pathogen and causative agent of nosocomial pneumonia especially in immunocompromised individuals in intensive care units. Recent advances to understand the contribution and function of A. baumannii virulence factors in its pathogenesis have begun to [...] Read more.
Acinetobacter baumannii has emerged as a significant opportunistic Gram-negative pathogen and causative agent of nosocomial pneumonia especially in immunocompromised individuals in intensive care units. Recent advances to understand the contribution and function of A. baumannii virulence factors in its pathogenesis have begun to elucidate how this bacterium interacts with immune cells and its interesting mechanisms for multi-antibiotic resistance. Taking advantage of the availability of the A. baumannii AB5075 transposon mutant library, we investigated the impact of the A. baumannii Clp genes, which encode for a chaperone-protease responsible for the degradation of misfolded proteins, on bacterial virulence in a model of pneumonia using C57BL/6 mice and survival within J774.16 macrophage-like cells. Clp-protease A. baumannii mutants exhibit decreased virulence in rodents, high phagocytic cell-mediated killing and reduced biofilm formation. Capsular staining showed evidence of encapsulation in A. baumannii AB5075 and Clp-mutant strains. Surprisingly, clpA and clpS mutants displayed irregular cell morphology, which may be important in the biofilm structural deficiencies observed in these strains. Interestingly, clpA showed apical-like growth, proliferation normally observed in filamentous fungi. These findings provide new information regarding A. baumannii pathogenesis and may be important for the development of therapies intended at reducing morbidity and mortality associated with this remarkable pathogen. Full article
(This article belongs to the Special Issue Microbial Interactions during Infection)
Show Figures

Figure 1

10 pages, 1591 KiB  
Article
Interspecies Metabolic Complementation in Cystic Fibrosis Pathogens via Purine Exchange
by Hafij Al Mahmud, Jiwasmika Baishya and Catherine A. Wakeman
Pathogens 2021, 10(2), 146; https://doi.org/10.3390/pathogens10020146 - 01 Feb 2021
Cited by 3 | Viewed by 2529
Abstract
Cystic fibrosis (CF) is a genetic disease frequently associated with chronic lung infections caused by a consortium of pathogens. It is common for auxotrophy (the inability to biosynthesize certain essential metabolites) to develop in clinical isolates of the dominant CF pathogen Pseudomonas aeruginosa [...] Read more.
Cystic fibrosis (CF) is a genetic disease frequently associated with chronic lung infections caused by a consortium of pathogens. It is common for auxotrophy (the inability to biosynthesize certain essential metabolites) to develop in clinical isolates of the dominant CF pathogen Pseudomonas aeruginosa, indicating that the CF lung environment is replete in various nutrients. Many of these nutrients are likely to come from the host tissues, but some may come from the surrounding polymicrobial community within the lungs of CF patients as well. To assess the feasibility of nutrient exchange within the polymicrobial community of the CF lung, we selected P. aeruginosa and Staphylococcus aureus, two of the most prevalent species found in the CF lung environment. By comparing the polymicrobial culture of wild-type strains relative to their purine auxotrophic counterparts, we were able to observe metabolic complementation occurring in both P. aeruginosa and S. aureus when grown with a purine-producing cross-species pair. While our data indicate that some of this complementation is likely derived from extracellular DNA freed by lysis of S. aureus by the highly competitive P. aeruginosa, the partial complementation of S. aureus purine deficiency by P. aeruginosa demonstrates that bidirectional nutrient exchange between these classic competitors is possible. Full article
(This article belongs to the Special Issue Microbial Interactions during Infection)
Show Figures

Figure 1

13 pages, 16783 KiB  
Article
The Relevance of IL-1-Signaling in the Protection against Gram-Positive Bacteria
by Angelina Midiri, Giuseppe Mancuso, Concetta Beninati, Elisabetta Gerace and Carmelo Biondo
Pathogens 2021, 10(2), 132; https://doi.org/10.3390/pathogens10020132 - 28 Jan 2021
Cited by 7 | Viewed by 1588
Abstract
Previous studies performed using a model of group B streptococcus (GBS)-induced peritoneal inflammation indicate that the interleukin-1 receptor (IL-1R) family plays an important role in the innate host defense against this encapsulated Gram-positive bacteria. Since the role of IL-1-dependent signaling in peritoneal infections [...] Read more.
Previous studies performed using a model of group B streptococcus (GBS)-induced peritoneal inflammation indicate that the interleukin-1 receptor (IL-1R) family plays an important role in the innate host defense against this encapsulated Gram-positive bacteria. Since the role of IL-1-dependent signaling in peritoneal infections induced by other Gram-positive bacteria is unknown, in the present study we sought to investigate the contribution of IL-1R signaling in host defenses against Streptococcus pyogenes (group A streptococcus or GAS) or Staphylococcus aureus, two frequent and global human Gram-positive extracellular pathogens. We analyzed here the outcome of GAS or S. aureus infection in IL-1R-deficient mice. After inoculated intraperitoneal (i.p.) inoculation with group A Streptococcus or S. aureus, all the wild-type (WT) control mice survived the challenge, while, respectively, 63% or 50% of IL-1-defective mice died. Lethality was due to the ability of both bacterial species to replicate and disseminate to the target organs of IL-1R-deficient mice. Moreover, the experimental results indicate that IL-1 signaling promotes the production of leukocyte attractant chemokines CXCL-1 and CXCL-2 and recruitment of neutrophils to bacterial infection sites. Accordingly, the reduced neutrophil recruitment in IL-1R-deficient mice was linked with decreased production of neutrophil chemokines. Collectively, our findings indicate that IL-1 signaling, as previously showed in host defense against GBS, plays a fundamental role also in controlling the progression and outcome of GAS or S. aureus disease. Full article
(This article belongs to the Special Issue Microbial Interactions during Infection)
Show Figures

Figure 1

10 pages, 1403 KiB  
Article
In Silico Analysis of Possible Interaction between Host Genomic Transcription Factors (TFs) and Zika Virus (ZikaSPH2015) Strain with Combinatorial Gene Regulation; Virus Versus Host—The Game Reloaded
by Massimiliano Chetta, Marina Tarsitano, Laura Vicari, Annalisa Saracino and Nenad Bukvic
Pathogens 2021, 10(1), 69; https://doi.org/10.3390/pathogens10010069 - 14 Jan 2021
Cited by 1 | Viewed by 1802
Abstract
In silico analysis is a promising approach for understanding biological events in complex diseases. Herein we report on the innovative computational workflow allowed to highlight new direct interactions between human transcription factors (TFs) and an entire genome of virus ZikaSPH2015 strain in order [...] Read more.
In silico analysis is a promising approach for understanding biological events in complex diseases. Herein we report on the innovative computational workflow allowed to highlight new direct interactions between human transcription factors (TFs) and an entire genome of virus ZikaSPH2015 strain in order to identify the occurrence of specific motifs on a genomic Zika Virus sequence that is able to bind and, therefore, sequester host’s TFs. The analysis pipeline was performed using different bioinformatics tools available online (free of charge). According to obtained results of this in silico analysis, it is possible to hypothesize that these TFs binding motifs might be able to explain the complex and heterogeneous phenotype presentation in Zika-virus-affected fetuses/newborns, as well as the less severe condition in adults. Moreover, the proposed in silico protocol identified thirty-three different TFs identical to the distribution of TFBSs (Transcription Factor Binding Sites) on ZikaSPH2015 strain, potentially able to influence genes and pathways with biological functions confirming that this approach could find potential answers on disease pathogenesis. Full article
(This article belongs to the Special Issue Microbial Interactions during Infection)
Show Figures

Figure 1

19 pages, 6283 KiB  
Article
Altered Salivary Microbiome in the Early Stage of HIV Infections among Young Chinese Men Who Have Sex with Men (MSM)
by Jin Li, Shenghua Chang, Haiying Guo, Yaoting Ji, Han Jiang, Lianguo Ruan and Minquan Du
Pathogens 2020, 9(11), 960; https://doi.org/10.3390/pathogens9110960 - 19 Nov 2020
Cited by 9 | Viewed by 2102 | Correction
Abstract
Human immunodeficiency virus (HIV) infections are spiking in Chinese young men who have sex with men (MSM). To explore alterations in the salivary microbiome and its correlation with demographic characteristics, CD4+ T cell count and viral load (VL) in HIV infections, samples of [...] Read more.
Human immunodeficiency virus (HIV) infections are spiking in Chinese young men who have sex with men (MSM). To explore alterations in the salivary microbiome and its correlation with demographic characteristics, CD4+ T cell count and viral load (VL) in HIV infections, samples of unstimulated whole saliva were analyzed by 16S rRNA gene sequencing using the Illumina MiSeq platform in 20 HIV newly infected patients before the initiation of antiretroviral therapy (ART) and at three and six months after, and in 20 age- and gender-paired healthy Chinese people. The results showed that the alpha diversity of salivary microbiota in HIV infections did not show differences from the healthy controls, but was reduced after six months under ART treatment. Comparative analysis revealed that Streptococcus was enriched in HIV-infected individuals, while Neisseria was enriched in the healthy control group. After effective ART, the salivary microbiota composition was not completely restored, although some microbiota recovered. In addition, we found Provotella_7, Neisseria and Haemophilus were correlated negatively with CD4+ T cell count, while Neisseria was correlated positively with VL. We conclude that HIV infections experience a dysbiosis of the salivary microbiome. The salivary microbiome test could be a substitute for the blood tests in the diagnosis and prognosis of diseases. Full article
(This article belongs to the Special Issue Microbial Interactions during Infection)
Show Figures

Figure 1

20 pages, 3795 KiB  
Article
Enterococcus faecalis Polymicrobial Interactions Facilitate Biofilm Formation, Antibiotic Recalcitrance, and Persistent Colonization of the Catheterized Urinary Tract
by Jordan R. Gaston, Marissa J. Andersen, Alexandra O. Johnson, Kirsten L. Bair, Christopher M. Sullivan, L. Beryl Guterman, Ashely N. White, Aimee L. Brauer, Brian S. Learman, Ana L. Flores-Mireles and Chelsie E. Armbruster
Pathogens 2020, 9(10), 835; https://doi.org/10.3390/pathogens9100835 - 13 Oct 2020
Cited by 30 | Viewed by 3820
Abstract
Indwelling urinary catheters are common in health care settings and can lead to catheter-associated urinary tract infection (CAUTI). Long-term catheterization causes polymicrobial colonization of the catheter and urine, for which the clinical significance is poorly understood. Through prospective assessment of catheter urine colonization, [...] Read more.
Indwelling urinary catheters are common in health care settings and can lead to catheter-associated urinary tract infection (CAUTI). Long-term catheterization causes polymicrobial colonization of the catheter and urine, for which the clinical significance is poorly understood. Through prospective assessment of catheter urine colonization, we identified Enterococcus faecalis and Proteus mirabilis as the most prevalent and persistent co-colonizers. Clinical isolates of both species successfully co-colonized in a murine model of CAUTI, and they were observed to co-localize on catheter biofilms during infection. We further demonstrate that P. mirabilis preferentially adheres to E. faecalis during biofilm formation, and that contact-dependent interactions between E. faecalis and P. mirabilis facilitate establishment of a robust biofilm architecture that enhances antimicrobial resistance for both species. E. faecalis may therefore act as a pioneer species on urinary catheters, establishing an ideal surface for persistent colonization by more traditional pathogens such as P. mirabilis. Full article
(This article belongs to the Special Issue Microbial Interactions during Infection)
Show Figures

Figure 1

14 pages, 4757 KiB  
Article
Inhibition of Mixed Biofilms of Candida albicans and Methicillin-Resistant Staphylococcus aureus by Positively Charged Silver Nanoparticles and Functionalized Silicone Elastomers
by Humberto H. Lara and Jose L. Lopez-Ribot
Pathogens 2020, 9(10), 784; https://doi.org/10.3390/pathogens9100784 - 25 Sep 2020
Cited by 21 | Viewed by 3061
Abstract
Both bacterial and fungal organisms display the ability to form biofilms; however, mixed bacterial/fungal biofilms are particularly difficult to control and eradicate. The opportunistic microbial pathogens Candida albicans and Staphylococcus aureus are among the most frequent causative agents of healthcare-acquired infections, and are [...] Read more.
Both bacterial and fungal organisms display the ability to form biofilms; however, mixed bacterial/fungal biofilms are particularly difficult to control and eradicate. The opportunistic microbial pathogens Candida albicans and Staphylococcus aureus are among the most frequent causative agents of healthcare-acquired infections, and are often co-isolated forming mixed biofilms, especially from contaminated catheters. These mixed species biofilms display a high level of antibiotic resistance; thus, these infections are challenging to treat resulting in excess morbidity and mortality. In the absence of effective conventional antibiotic treatments, nanotechnology-based approaches represent a promising alternative for the treatment of highly recalcitrant polymicrobial biofilm infections. Our group has previously reported on the activity of pure positively charged silver nanoparticles synthesized by a novel microwave technique against single-species biofilms of C. albicans and S. aureus. Here, we have expanded our observations to demonstrate that that silver nanoparticles display dose-dependent activity against dual-species C. albicans/S. aureus biofilms. Moreover, the same nanoparticles were used to functionalize catheter materials, leading to the effective inhibition of the mixed fungal/bacterial biofilms. Overall, our results indicate the potent activity of silver nanoparticles against these cross-kingdom biofilms. More studies are warranted to examine the ability of functionalized catheters in the prevention of catheter-related bloodstream infections. Full article
(This article belongs to the Special Issue Microbial Interactions during Infection)
Show Figures

Figure 1

15 pages, 3214 KiB  
Article
The Examination of Viral Characteristics of HIV-1 CRF07_BC and Its Potential Interaction with Extracellular Galectin-3
by Chih-Yen Lin, Wen-Hung Wang, Szu-Wei Huang, Chun-Sheng Yeh, Ruei-Yu Yuan, Zih-Syuan Yang, Aspiro Nayim Urbina, Sung-Pin Tseng, Po-Liang Lu, Yen-Hsu Chen and Sheng-Fan Wang
Pathogens 2020, 9(6), 425; https://doi.org/10.3390/pathogens9060425 - 29 May 2020
Cited by 4 | Viewed by 2215
Abstract
HIV-1 CRF07_BC is a B’ and C subtype recombinant emerging virus and many of its viral characteristics remain unclear. Galectin-3 (Gal3) is a β-galactose binding lectin that has been reported as a pattern recognition receptor (PRR) and is known to mediate adhesion between [...] Read more.
HIV-1 CRF07_BC is a B’ and C subtype recombinant emerging virus and many of its viral characteristics remain unclear. Galectin-3 (Gal3) is a β-galactose binding lectin that has been reported as a pattern recognition receptor (PRR) and is known to mediate adhesion between cells and microbes. This study aims to examine the viral characteristics of HIV-1 CRF07_BC virus and the role of extracellular galectin-3 in HIV-1 CRF07_BC infection. A total of 28 HIV-1+ injecting drug users (IDUs) were recruited and 24 (85.7%) were identified as HIV-1 CRF07_BC. Results indicate that significant higher serum galectin-3 was measured in CRF07_BC infected patients and CRF07_BC infection triggered significant galectin-3 expression (p < 0.01). Viral characteristics demonstrate that CRF07_BC virions display a higher level of envelope gp120 spikes. The virus infectivity assay demonstrated that co-treatment with galectin-3 significantly promoted CRF07_BC attachment and internalization (p < 0.01). A co-immunoprecipitation assay showed that pulldown galectin-3 co-precipitated both CD4 and gp120 proteins. Results from an enzyme-linked immunosorbent assay (ELISA) indicate that the galectin-3 promoting effect occurs through enhancement of the interaction between gp120 and CD4. This study suggests that CRF07_BC was predominant in HIV-1+ IDUs and CRF07_BC utilized extracellular galectin-3 to enhance its infectivity via stabilization of the gp120-CD4 interaction. Full article
(This article belongs to the Special Issue Microbial Interactions during Infection)
Show Figures

Figure 1

Review

Jump to: Research, Other

29 pages, 2169 KiB  
Review
Current State and Promising Opportunities on Pharmaceutical Approaches in the Treatment of Polymicrobial Diseases
by Sartini Sartini, Andi Dian Permana, Saikat Mitra, Abu Montakim Tareq, Emil Salim, Islamudin Ahmad, Harapan Harapan, Talha Bin Emran and Firzan Nainu
Pathogens 2021, 10(2), 245; https://doi.org/10.3390/pathogens10020245 - 20 Feb 2021
Cited by 13 | Viewed by 4227
Abstract
In recent years, the emergence of newly identified acute and chronic infectious disorders caused by diverse combinations of pathogens, termed polymicrobial diseases, has had catastrophic consequences for humans. Antimicrobial agents have been clinically proven to be effective in the pharmacological treatment of polymicrobial [...] Read more.
In recent years, the emergence of newly identified acute and chronic infectious disorders caused by diverse combinations of pathogens, termed polymicrobial diseases, has had catastrophic consequences for humans. Antimicrobial agents have been clinically proven to be effective in the pharmacological treatment of polymicrobial diseases. Unfortunately, an increasing trend in the emergence of multi-drug-resistant pathogens and limited options for delivery of antimicrobial drugs might seriously impact humans’ efforts to combat polymicrobial diseases in the coming decades. New antimicrobial agents with novel mechanism(s) of action and new pharmaceutical formulations or delivery systems to target infected sites are urgently required. In this review, we discuss the prospective use of novel antimicrobial compounds isolated from natural products to treat polymicrobial infections, mainly via mechanisms related to inhibition of biofilm formation. Drug-delivery systems developed to deliver antimicrobial compounds to both intracellular and extracellular pathogens are discussed. We further discuss the effectiveness of several biofilm-targeted delivery strategies to eliminate polymicrobial biofilms. At the end, we review the applications and promising opportunities for various drug-delivery systems, when compared to conventional antimicrobial therapy, as a pharmacological means to treat polymicrobial diseases. Full article
(This article belongs to the Special Issue Microbial Interactions during Infection)
Show Figures

Figure 1

34 pages, 2135 KiB  
Review
Interplay between ESKAPE Pathogens and Immunity in Skin Infections: An Overview of the Major Determinants of Virulence and Antibiotic Resistance
by Gustavo Henrique Rodrigues Vale de Macedo, Gabrielle Damasceno Evangelista Costa, Elane Rodrigues Oliveira, Glauciane Viera Damasceno, Juliana Silva Pereira Mendonça, Lucas dos Santos Silva, Vitor Lopes Chagas, José Manuel Noguera Bazán, Amanda Silva dos Santos Aliança, Rita de Cássia Mendonça de Miranda, Adrielle Zagmignan, Andrea de Souza Monteiro and Luís Cláudio Nascimento da Silva
Pathogens 2021, 10(2), 148; https://doi.org/10.3390/pathogens10020148 - 02 Feb 2021
Cited by 28 | Viewed by 4670
Abstract
The skin is the largest organ in the human body, acting as a physical and immunological barrier against pathogenic microorganisms. The cutaneous lesions constitute a gateway for microbial contamination that can lead to chronic wounds and other invasive infections. Chronic wounds are considered [...] Read more.
The skin is the largest organ in the human body, acting as a physical and immunological barrier against pathogenic microorganisms. The cutaneous lesions constitute a gateway for microbial contamination that can lead to chronic wounds and other invasive infections. Chronic wounds are considered as serious public health problems due the related social, psychological and economic consequences. The group of bacteria known as ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter sp.) are among the most prevalent bacteria in cutaneous infections. These pathogens have a high level of incidence in hospital environments and several strains present phenotypes of multidrug resistance. In this review, we discuss some important aspects of skin immunology and the involvement of ESKAPE in wound infections. First, we introduce some fundamental aspects of skin physiology and immunology related to cutaneous infections. Following this, the major virulence factors involved in colonization and tissue damage are highlighted, as well as the most frequently detected antimicrobial resistance genes. ESKAPE pathogens express several virulence determinants that overcome the skin’s physical and immunological barriers, enabling them to cause severe wound infections. The high ability these bacteria to acquire resistance is alarming, particularly in the hospital settings where immunocompromised individuals are exposed to these pathogens. Knowledge about the virulence and resistance markers of these species is important in order to develop new strategies to detect and treat their associated infections. Full article
(This article belongs to the Special Issue Microbial Interactions during Infection)
Show Figures

Figure 1

12 pages, 1052 KiB  
Review
Clinical Implications of Polymicrobial Synergism Effects on Antimicrobial Susceptibility
by William Little, Caroline Black and Allie Clinton Smith
Pathogens 2021, 10(2), 144; https://doi.org/10.3390/pathogens10020144 - 01 Feb 2021
Cited by 16 | Viewed by 4536
Abstract
With the development of next generation sequencing technologies in recent years, it has been demonstrated that many human infectious processes, including chronic wounds, cystic fibrosis, and otitis media, are associated with a polymicrobial burden. Research has also demonstrated that polymicrobial infections tend to [...] Read more.
With the development of next generation sequencing technologies in recent years, it has been demonstrated that many human infectious processes, including chronic wounds, cystic fibrosis, and otitis media, are associated with a polymicrobial burden. Research has also demonstrated that polymicrobial infections tend to be associated with treatment failure and worse patient prognoses. Despite the importance of the polymicrobial nature of many infection states, the current clinical standard for determining antimicrobial susceptibility in the clinical laboratory is exclusively performed on unimicrobial suspensions. There is a growing body of research demonstrating that microorganisms in a polymicrobial environment can synergize their activities associated with a variety of outcomes, including changes to their antimicrobial susceptibility through both resistance and tolerance mechanisms. This review highlights the current body of work describing polymicrobial synergism, both inter- and intra-kingdom, impacting antimicrobial susceptibility. Given the importance of polymicrobial synergism in the clinical environment, a new system of determining antimicrobial susceptibility from polymicrobial infections may significantly impact patient treatment and outcomes. Full article
(This article belongs to the Special Issue Microbial Interactions during Infection)
Show Figures

Figure 1

11 pages, 2409 KiB  
Review
The Impact of Intraspecies and Interspecies Bacterial Interactions on Disease Outcome
by Jiwasmika Baishya, Karishma Bisht, Jeanette N. Rimbey, Kiddist D. Yihunie, Shariful Islam, Hafij Al Mahmud, Jayc E. Waller and Catherine A. Wakeman
Pathogens 2021, 10(2), 96; https://doi.org/10.3390/pathogens10020096 - 21 Jan 2021
Cited by 8 | Viewed by 3999
Abstract
The human microbiota is an array of microorganisms known to interact with the host and other microbes. These interactions can be competitive, as microbes must adapt to host- and microorganism-related stressors, thus producing toxic molecules, or cooperative, whereby microbes survive by maintaining homeostasis [...] Read more.
The human microbiota is an array of microorganisms known to interact with the host and other microbes. These interactions can be competitive, as microbes must adapt to host- and microorganism-related stressors, thus producing toxic molecules, or cooperative, whereby microbes survive by maintaining homeostasis with the host and host-associated microbial communities. As a result, these microbial interactions shape host health and can potentially result in disease. In this review, we discuss these varying interactions across microbial species, their positive and negative effects, the therapeutic potential of these interactions, and their implications on our knowledge of human well-being. Full article
(This article belongs to the Special Issue Microbial Interactions during Infection)
Show Figures

Figure 1

15 pages, 1009 KiB  
Review
Microbial Etiology and Prevention of Dental Caries: Exploiting Natural Products to Inhibit Cariogenic Biofilms
by Xiuqin Chen, Eric Banan-Mwine Daliri, Namhyeon Kim, Jong-Rae Kim, Daesang Yoo and Deog-Hwan Oh
Pathogens 2020, 9(7), 569; https://doi.org/10.3390/pathogens9070569 - 14 Jul 2020
Cited by 114 | Viewed by 14394
Abstract
Dental caries is one of the most common microbe-mediated oral diseases in human beings. At present, the accepted etiology of caries is based on a four-factor theory that includes oral microorganisms, oral environment, host, and time. Excessive exposure to dietary carbohydrates leads to [...] Read more.
Dental caries is one of the most common microbe-mediated oral diseases in human beings. At present, the accepted etiology of caries is based on a four-factor theory that includes oral microorganisms, oral environment, host, and time. Excessive exposure to dietary carbohydrates leads to the accumulation of acid-producing and acid-resistant microorganisms in the mouth. Dental caries is driven by dysbiosis of the dental biofilm adherent to the enamel surface. Effective preventive methods include inhibiting the cariogenic microorganisms, treatment with an anti-biofilm agent, and sugar intake control. The goal is to reduce the total amount of biofilm or the levels of specific pathogens. Natural products could be recommended for preventing dental caries, since they may possess fewer side effects in comparison with synthetic antimicrobials. Herein, the mechanisms of oral microbial community development and functional specialization are discussed. We highlight the application of widely explored natural products in the last five years for their ability to inhibit cariogenic microorganisms. Full article
(This article belongs to the Special Issue Microbial Interactions during Infection)
Show Figures

Figure 1

Other

Jump to: Research, Review

1 pages, 2834 KiB  
Correction
Correction: Li et al. Altered Salivary Microbiome in the Early Stage of HIV Infections among Young Chinese Men Who Have Sex with Men (MSM). Pathogens 2020, 9, 960
by Jin Li, Shenghua Chang, Haiying Guo, Yaoting Ji, Han Jiang, Lianguo Ruan and Minquan Du
Pathogens 2022, 11(7), 785; https://doi.org/10.3390/pathogens11070785 - 11 Jul 2022
Viewed by 821
Abstract
In the original publication [...] Full article
(This article belongs to the Special Issue Microbial Interactions during Infection)
Show Figures

Figure 1

9 pages, 442 KiB  
Case Report
Escherichia coli Strains with Virulent Factors Typical for Uropathogens were Isolated from Sinuses from Patients with Chronic Rhinosinusitis—Case Report
by Beata Krawczyk, Michał Michalik, Magdalena Fordon, Magdalena Wysocka, Alfred Samet and Bogdan Nowicki
Pathogens 2020, 9(5), 318; https://doi.org/10.3390/pathogens9050318 - 25 Apr 2020
Cited by 4 | Viewed by 3875
Abstract
Escherichia coli were isolated from three patients with chronic rhinosinusitis (CRS) by intraoperative sinus tissue biopsy. Taking into account the unusual replicative niche and previous treatment failures, it was decided to focus on the virulence and drug resistance of these bacteria. The strains [...] Read more.
Escherichia coli were isolated from three patients with chronic rhinosinusitis (CRS) by intraoperative sinus tissue biopsy. Taking into account the unusual replicative niche and previous treatment failures, it was decided to focus on the virulence and drug resistance of these bacteria. The strains turned out to be multi-sensitive, but the rich virulence factors profile of bacteria typical for phylogenetic group B2 deserved attention. Tests were carried out for the presence of 32 genes using the PCR method. Particularly noteworthy are the toxins Cnf-1, HlyA, Usp—an extensive iron uptake system (enterobactin, salmochelin, yersiniabactin and outer membrane hemin receptor ChuA)—SPATE autotransporters such as vat and pic, Ag43 autoaggregative protein—important for biofilm formation—and TosA/B which enhance the fitness of E.coli. All these virulence factors are identified predominantly in UPEC strains and provide a fitness advantage during colonization of the sinuses. Patients with CRS should be asked for past or present UTI. The specific virulence factors of E. coli that facilitate the colonization of the GI tract and urinary tract may also favor the colonization of a new ecological niche (sinuses) as a result of microbial imbalance or dysbiosis. Full article
(This article belongs to the Special Issue Microbial Interactions during Infection)
Show Figures

Figure 1

Back to TopTop