ijms-logo

Journal Browser

Journal Browser

Molecular Mechanisms of Host-Pathogen Interaction

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Biology".

Deadline for manuscript submissions: closed (31 December 2021) | Viewed by 60933

Special Issue Editors


E-Mail Website
Guest Editor
M2iSH (Microbes, intestine, inflammation and Susceptibility of the Host), Inserm U1071, INRA USC 2018, University of Clermont Auvergne, 63000 Clermont–Ferrand, France
Interests: inflammatory bowel diseases; colorectal cancer; host-pathogen interaction; mucosal immune response; intestinal microbiota; microRNA; autophagy; exosomes

E-Mail Website
Guest Editor
Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH); Université Clermont Auvergne/Inserm U1071, USC-INRA 2018, Clermont-Ferrand, F-63001, France
Interests: epigenetics; nutrition; mechanisms of bacterial invasion; pathobiont; host-response to bacteria

E-Mail Website
Guest Editor
Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH) UMR 1071 Inserm/Université Clermont Auvergne, USC-INRA 2018, CRNH Auvergne, F-63000 Clermont-Ferrand, France
Interests: immune response; pathobiont E. coli; nutrition; microbiota

Special Issue Information

Dear Colleagues,

The interaction between host and bacterial pathogens is emerging as a key area in infectious disease research. Pathogens have evolved multiple mechanisms to combat the host defense as they establish infections. These could include, among others, the capacity of bacterial pathogens to overcome protective host barriers, to regulate host autophagy to avoid their recognition and/or to promote their own replication, to induce epigenetic modulation of the host and to subvert the immune system, which may lead to exacerbated inflammation. In addition, the extracellular vesicles secreted by the host cells during pathogen infection could modify susceptibility to infection by transferring their composition from cell-to-cell. This Special Issue will take an interest in manuscripts that report current data or summarize the current understanding of the complex interplay between the host and microorganisms, and the cell-to-cell communication during host–pathogen infection. Interdisciplinary reviews and research articles of various fields, such as microbiology, immunology, cell biology, genetics, and ecology, are welcome.

Dr. Hang Thi Thu Nguyen
Dr. Jéremy Jéremy Denizot
Dr. Elisabeth Billard
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. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. 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

  • Dysbiosis
  • Bacterial infection
  • Bacteria–epithelium interaction
  • Autophagy
  • Epigenetics
  • DNA methylation
  • Histone post-translational modification
  • Nutrition and bacterial infection
  • Immunomodulation
  • Exosomes

Published Papers (17 papers)

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

Research

Jump to: Review, Other

23 pages, 3618 KiB  
Article
Multiomics Analysis of Endocytosis upon HBV Infection and Identification of SCAMP1 as a Novel Host Restriction Factor against HBV Replication
by Tanzeel Yousaf, Yuting Sun, Wajeeha Naz, Yang Liu, Jiaqi Xu, Sen Yuan, Kangwei Wu, Min Wang, Jun Wang, Mingxiong Guo and Guihong Sun
Int. J. Mol. Sci. 2022, 23(4), 2211; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23042211 - 17 Feb 2022
Cited by 2 | Viewed by 2293
Abstract
Hepatitis B virus (HBV) infection remains a major global health problem and the primary cause of cirrhosis and hepatocellular carcinoma (HCC). HBV intrusion into host cells is prompted by virus–receptor interactions in clathrin-mediated endocytosis. Here, we report a comprehensive view of the cellular [...] Read more.
Hepatitis B virus (HBV) infection remains a major global health problem and the primary cause of cirrhosis and hepatocellular carcinoma (HCC). HBV intrusion into host cells is prompted by virus–receptor interactions in clathrin-mediated endocytosis. Here, we report a comprehensive view of the cellular endocytosis-associated transcriptome, proteome and ubiquitylome upon HBV infection. In this study, we quantified 273 genes in the transcriptome and 190 endocytosis-associated proteins in the proteome by performing multi-omics analysis. We further identified 221 Lys sites in 77 endocytosis-associated ubiquitinated proteins. A weak negative correlation was observed among endocytosis-associated transcriptome, proteome and ubiquitylome. We found 33 common differentially expressed genes (DEGs), differentially expressed proteins (DEPs), and Kub-sites. Notably, we reported the HBV-induced ubiquitination change of secretory carrier membrane protein (SCAMP1) for the first time, differentially expressed across all three omics data sets. Overexpression of SCAMP1 efficiently inhibited HBV RNAs/pgRNA and secreted viral proteins, whereas knockdown of SCAMP1 significantly increased viral production. Mechanistically, the EnhI/XP, SP1, and SP2 promoters were inhibited by SCAMP1, which accounts for HBV X and S mRNA inhibition. Overall, our study unveils the previously unknown role of SCAMP1 in viral replication and HBV pathogenesis and provides cumulative and novel information for a better understanding of endocytosis in response to HBV infection. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Host-Pathogen Interaction)
Show Figures

Figure 1

21 pages, 3122 KiB  
Article
Pseudomonas aeruginosa Triggered Exosomal Release of ADAM10 Mediates Proteolytic Cleavage in Trans
by Ahmad Aljohmani, Bastian Opitz, Markus Bischoff and Daniela Yildiz
Int. J. Mol. Sci. 2022, 23(3), 1259; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23031259 - 23 Jan 2022
Cited by 4 | Viewed by 2743
Abstract
Pneumonia is a life-threatening disease often caused by infection with Streptococcus pneumoniae and Pseudomonas aeruginosa. Many of the mediators (e.g., TNF, IL-6R) and junction molecules (e.g., E-cadherin) orchestrating inflammatory cell recruitment and loss of barrier integrity are proteolytically cleaved through a disintegrin [...] Read more.
Pneumonia is a life-threatening disease often caused by infection with Streptococcus pneumoniae and Pseudomonas aeruginosa. Many of the mediators (e.g., TNF, IL-6R) and junction molecules (e.g., E-cadherin) orchestrating inflammatory cell recruitment and loss of barrier integrity are proteolytically cleaved through a disintegrin and metalloproteinases (ADAMs). We could show by Western blot, surface expression analysis and measurement of proteolytic activity in cell-based assays, that ADAM10 in epithelial cells is upregulated and activated upon infection with Pseudomonas aeruginosa and Exotoxin A (ExoA), but not upon infection with Streptococcus pneumoniae. Targeting ADAM10 by pharmacological inhibition or gene silencing, we demonstrated that this activation was critical for cleavage of E-cadherin and modulated permeability and epithelial integrity. Stimulation with heat-inactivated bacteria revealed that the activation was based on the toxin repertoire rather than the interaction with the bacterial particle itself. Furthermore, calcium imaging experiments showed that the ExoA action was based on the induction of calcium influx. Investigating the extracellular vesicles and their proteolytic activity, we could show that Pseudomonas aeruginosa triggered exosomal release of ADAM10 and proteolytic cleavage in trans. This newly described mechanism could constitute an essential mechanism causing systemic inflammation in patients suffering from Pseudomonas aeruginosa-induced pneumonia stimulating future translational studies. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Host-Pathogen Interaction)
Show Figures

Figure 1

16 pages, 2190 KiB  
Article
Investigating Pathogenicity and Virulence of Staphylococcus pettenkoferi: An Emerging Pathogen
by Nour Ahmad-Mansour, Lucile Plumet, Sylvaine Huc-Brandt, Chloé Magnan, Alex Yahiaoui-Martinez, Karima Kissa, Alix Pantel, Jean-Philippe Lavigne and Virginie Molle
Int. J. Mol. Sci. 2021, 22(24), 13614; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222413614 - 19 Dec 2021
Cited by 10 | Viewed by 3807
Abstract
Staphylococcus pettenkoferi is a coagulase-negative Staphylococcus identified in 2002 that has been implicated in human diseases as an opportunistic pathogenic bacterium. Its multiresistant character is becoming a major health problem, yet the pathogenicity of S. pettenkoferi is poorly characterized. In this study, the [...] Read more.
Staphylococcus pettenkoferi is a coagulase-negative Staphylococcus identified in 2002 that has been implicated in human diseases as an opportunistic pathogenic bacterium. Its multiresistant character is becoming a major health problem, yet the pathogenicity of S. pettenkoferi is poorly characterized. In this study, the pathogenicity of a S. pettenkoferi clinical isolate from diabetic foot osteomyelitis was compared with a Staphylococcus aureus strain in various in vitro and in vivo experiments. Growth kinetics were compared against S. aureus, and bacteria survival was assessed in the RAW 264.7 murine macrophage cell line, the THP-1 human leukemia monocytic cell line, and the HaCaT human keratinocyte cell line. Ex vivo analysis was performed in whole blood survival assays and in vivo assays via the infection model of zebrafish embryos. Moreover, whole-genome analysis was performed. Our results show that S. pettenkoferi was able to survive in human blood, human keratinocytes, murine macrophages, and human macrophages. S. pettenkoferi demonstrated its virulence by causing substantial embryo mortality in the zebrafish model. Genomic analysis revealed virulence factors such as biofilm-encoding genes (e.g., icaABCD; rsbUVW) and regulator-encoding genes (e.g., agr, mgrA, sarA, saeS) well characterized in S. aureus. This study thus advances the knowledge of this under-investigated pathogen and validates the zebrafish infection model for this bacterium. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Host-Pathogen Interaction)
Show Figures

Figure 1

17 pages, 3367 KiB  
Article
HPE1, an Effector from Zebra Chip Pathogen Interacts with Tomato Proteins and Perturbs Ubiquitinated Protein Accumulation
by Chia-Cheng Kan, Azucena Mendoza-Herrera, Julien Levy, J. Joe Hull, Jeffery A. Fabrick and Cecilia Tamborindeguy
Int. J. Mol. Sci. 2021, 22(16), 9003; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22169003 - 20 Aug 2021
Cited by 5 | Viewed by 2385
Abstract
The gram-negative bacterial genus Liberibacter includes economically important pathogens, such as ‘Candidatus Liberibacter asiaticus’ that cause citrus greening disease (or Huanglongbing, HLB) and ‘Ca. Liberibacter solanacearum’ (Lso) that cause zebra chip disease in potato. Liberibacter pathogens are fastidious bacteria transmitted by [...] Read more.
The gram-negative bacterial genus Liberibacter includes economically important pathogens, such as ‘Candidatus Liberibacter asiaticus’ that cause citrus greening disease (or Huanglongbing, HLB) and ‘Ca. Liberibacter solanacearum’ (Lso) that cause zebra chip disease in potato. Liberibacter pathogens are fastidious bacteria transmitted by psyllids. Pathogen manipulation of the host’ and vector’s immune system for successful colonization is hypothesized to be achieved by Sec translocon-dependent effectors (SDE). In previous work, we identified hypothetical protein effector 1 (HPE1), an SDE from Lso, that acts as a suppressor of the plant’s effector-triggered immunity (ETI)-like response. In this study, using a yeast two-hybrid system, we identify binding interactions between tomato RAD23 proteins and HPE1. We further show that HPE1 interacts with RAD23 in both nuclear and cytoplasmic compartments in planta. Immunoblot assays show that HPE1 is not ubiquitinated in the plant cell, but rather the expression of HPE1 induced the accumulation of other ubiquitinated proteins. A similar accumulation of ubiquitinated proteins is also observed in Lso infected tomato plants. Finally, earlier colonization and symptom development following Lso haplotype B infection are observed in HPE1 overexpressing plants compared to wild-type plants. Overall, our results suggest that HPE1 plays a role in virulence in Lso pathogenesis, possibly by perturbing the ubiquitin-proteasome system via direct interaction with the ubiquitin-like domain of RAD23 proteins. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Host-Pathogen Interaction)
Show Figures

Figure 1

17 pages, 4016 KiB  
Article
Yersiniabactin Siderophore of Crohn’s Disease-Associated Adherent-Invasive Escherichia coli Is Involved in Autophagy Activation in Host Cells
by Guillaume Dalmasso, Hang Thi Thu Nguyen, Tiphanie Faïs, Sébastien Massier, Caroline Chevarin, Emilie Vazeille, Nicolas Barnich, Julien Delmas and Richard Bonnet
Int. J. Mol. Sci. 2021, 22(7), 3512; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22073512 - 29 Mar 2021
Cited by 6 | Viewed by 2282
Abstract
Background: Adherent-invasive Escherichia coli (AIEC) have been implicated in the etiology of Crohn’s disease. The AIEC reference strain LF82 possesses a pathogenicity island similar to the high pathogenicity island of Yersinia spp., which encodes the yersiniabactin siderophore required for iron uptake and growth [...] Read more.
Background: Adherent-invasive Escherichia coli (AIEC) have been implicated in the etiology of Crohn’s disease. The AIEC reference strain LF82 possesses a pathogenicity island similar to the high pathogenicity island of Yersinia spp., which encodes the yersiniabactin siderophore required for iron uptake and growth of the bacteria in iron-restricted environment. Here, we investigated the role of yersiniabactin during AIEC infection. Methods: Intestinal epithelial T84 cells and CEABAC10 transgenic mice were infected with LF82 or its mutants deficient in yersiniabactin expression. Autophagy was assessed by Western blot analysis for p62 and LC3-II expression. Results: Loss of yersiniabactin decreased the growth of LF82 in competitive conditions, reducing the ability of LF82 to adhere to and invade T84 cells and to colonize the intestinal tract of CEABAC10 mice. However, yersiniabactin deficiency increased LF82 intracellular replication. Mechanistically, a functional yersiniabactin is necessary for LF82-induced expression of HIF-1α, which is implicated in autophagy activation in infected cells. Conclusion: Our study highlights a novel role for yersiniabactin siderophore in AIEC–host interaction. Indeed, yersiniabactin, which is an advantage for AIEC to growth in a competitive environment, could be a disadvantage for the bacteria as it activates autophagy, a key host defense mechanism, leading to bacterial clearance. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Host-Pathogen Interaction)
Show Figures

Figure 1

15 pages, 2440 KiB  
Article
Consumption of Select Dietary Emulsifiers Exacerbates the Development of Spontaneous Intestinal Adenoma
by Emilie Viennois and Benoit Chassaing
Int. J. Mol. Sci. 2021, 22(5), 2602; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22052602 - 05 Mar 2021
Cited by 15 | Viewed by 2515
Abstract
Inflammation is a well-characterized critical driver of gastrointestinal cancers. Previous findings have shown that intestinal low-grade inflammation can be promoted by the consumption of select dietary emulsifiers, ubiquitous component of processed foods which alter the composition and function of the gut microbiota. Using [...] Read more.
Inflammation is a well-characterized critical driver of gastrointestinal cancers. Previous findings have shown that intestinal low-grade inflammation can be promoted by the consumption of select dietary emulsifiers, ubiquitous component of processed foods which alter the composition and function of the gut microbiota. Using a model of colitis-associated cancer, we previously reported that consumption of the dietary emulsifiers carboxymethylcellulose or polysorbate-80 exacerbated colonic tumor development. Here, we investigate the impact of dietary emulsifiers consumption on cancer initiation and progression in a genetical model of intestinal adenomas. In APCmin mice, we observed that dietary emulsifiers consumption enhanced small-intestine tumor development in a way that appeared to be independent of chronic intestinal inflammation but rather associated with emulsifiers’ impact on the proliferative status of the intestinal epithelium as well as on intestinal microbiota composition in both male and female mice. Overall, our findings further support the hypothesis that emulsifier consumption may be a new modifiable risk factor for colorectal cancer (CRC) and that alterations in host–microbiota interactions can favor gastrointestinal carcinogenesis in individuals with a genetical predisposition to such disorders. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Host-Pathogen Interaction)
Show Figures

Figure 1

27 pages, 4343 KiB  
Article
Structural Insights into the Interactions of Candidal Enolase with Human Vitronectin, Fibronectin and Plasminogen
by Dorota Satala, Grzegorz Satala, Justyna Karkowska-Kuleta, Michal Bukowski, Anna Kluza, Maria Rapala-Kozik and Andrzej Kozik
Int. J. Mol. Sci. 2020, 21(21), 7843; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21217843 - 22 Oct 2020
Cited by 24 | Viewed by 3019
Abstract
Significant amounts of enolase—a cytosolic enzyme involved in the glycolysis pathway—are exposed on the cell surface of Candida yeast. It has been hypothesized that this exposed enolase form contributes to infection-related phenomena such as fungal adhesion to human tissues, and the activation of [...] Read more.
Significant amounts of enolase—a cytosolic enzyme involved in the glycolysis pathway—are exposed on the cell surface of Candida yeast. It has been hypothesized that this exposed enolase form contributes to infection-related phenomena such as fungal adhesion to human tissues, and the activation of fibrinolysis and extracellular matrix degradation. The aim of the present study was to characterize, in structural terms, the protein-protein interactions underlying these moonlighting functions of enolase. The tight binding of human vitronectin, fibronectin and plasminogen by purified C. albicans and C. tropicalis enolases was quantitatively analyzed by surface plasmon resonance measurements, and the dissociation constants of the formed complexes were determined to be in the 10−7–10−8 M range. In contrast, the binding of human proteins by the S.cerevisiae enzyme was much weaker. The chemical cross-linking method was used to map the sites on enolase molecules that come into direct contact with human proteins. An internal motif 235DKAGYKGKVGIAMDVASSEFYKDGK259 in C. albicans enolase was suggested to contribute to the binding of all three human proteins tested. Models for these interactions were developed and revealed the sites on the enolase molecule that bind human proteins, extensively overlap for these ligands, and are well-separated from the catalytic activity center. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Host-Pathogen Interaction)
Show Figures

Figure 1

16 pages, 3022 KiB  
Article
In the Model Host Caenorhabditis elegans, Sphingosine-1-Phosphate-Mediated Signaling Increases Immunity toward Human Opportunistic Bacteria
by Kiho Lee, Iliana Escobar, Yeeun Jang, Wooseong Kim, Frederick M. Ausubel and Eleftherios Mylonakis
Int. J. Mol. Sci. 2020, 21(21), 7813; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21217813 - 22 Oct 2020
Cited by 5 | Viewed by 2795
Abstract
Sphingosine-1-phophate (S1P) is a sphingolipid-derived signaling molecule that controls diverse cellular functions including cell growth, homeostasis, and stress responses. In a variety of metazoans, cytosolic S1P is transported into the extracellular space where it activates S1P receptors in a concentration-dependent manner. In the [...] Read more.
Sphingosine-1-phophate (S1P) is a sphingolipid-derived signaling molecule that controls diverse cellular functions including cell growth, homeostasis, and stress responses. In a variety of metazoans, cytosolic S1P is transported into the extracellular space where it activates S1P receptors in a concentration-dependent manner. In the free-living nematode Caenorhabditis elegans, the spin-2 gene, which encodes a S1P transporter, is activated during Gram-positive or Gram-negative bacterial infection of the intestine. However, the role during infection of spin-2 and three additional genes in the C. elegans genome encoding other putative S1P transporters has not been elucidated. Here, we report an evolutionally conserved function for S1P and a non-canonical role for S1P transporters in the C. elegans immune response to bacterial pathogens. We found that mutations in the sphingosine kinase gene (sphk-1) or in the S1P transporter genes spin-2 or spin-3 decreased nematode survival after infection with Pseudomonas aeruginosa or Enterococcus faecalis. In contrast to spin-2 and spin-3, mutating spin-1 leads to an increase in resistance to P. aeruginosa. Consistent with these results, when wild-type C. elegans were supplemented with extracellular S1P, we found an increase in their lifespan when challenged with P. aeruginosa and E. faecalis. In comparison, spin-2 and spin-3 mutations suppressed the ability of S1P to rescue the worms from pathogen-mediated killing, whereas the spin-1 mutation had no effect on the immune-enhancing activity of S1P. S1P demonstrated no antimicrobial activity toward P. aeruginosa and Escherichia coli and only minimal activity against E. faecalis MMH594 (40 µM). These data suggest that spin-2 and spin-3, on the one hand, and spin-1, on the other hand, transport S1P across cellular membranes in opposite directions. Finally, the immune modulatory effect of S1P was diminished in C. eleganssek-1 and pmk-1 mutants, suggesting that the immunomodulatory effects of S1P are mediated by the p38 MAPK signaling pathway. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Host-Pathogen Interaction)
Show Figures

Figure 1

15 pages, 2506 KiB  
Article
Investigation of a Staphylococcus argenteus Strain Involved in a Chronic Prosthetic-Joint Infection
by Alan Diot, Virginie Dyon-Tafani, Marine Bergot, Jason Tasse, Patricia Martins-Simões, Jérôme Josse, Florent Valour and Frédéric Laurent
Int. J. Mol. Sci. 2020, 21(17), 6245; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21176245 - 28 Aug 2020
Cited by 7 | Viewed by 2646
Abstract
Staphylococcus argenteus is an emerging species responsible for infections comparable to those induced by Staphylococcus aureus. It has been involved in few chronic or persistent infections so far. In this study, we described a case of a persistent prosthetic-joint infection (PJI) affecting [...] Read more.
Staphylococcus argenteus is an emerging species responsible for infections comparable to those induced by Staphylococcus aureus. It has been involved in few chronic or persistent infections so far. In this study, we described a case of a persistent prosthetic-joint infection (PJI) affecting a young woman. We investigated in vitro the virulence traits of the incriminated S. argenteus strain (bone cell invasion, biofilm formation and induction of inflammation) and analyzed its genome, in comparison with two other strains of S. argenteus and two S. aureus isolates. It appeared that this S. argenteus PJI strain combined biofilm formation, osteoblast invasion and intracellular persistence abilities together with genes potentially involved in the escape of the host immune defenses, which might explain the chronicization of the infection. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Host-Pathogen Interaction)
Show Figures

Figure 1

Review

Jump to: Research, Other

18 pages, 1313 KiB  
Review
Involvement of the Innate Immune System in the Pathogenesis of Chronic Obstructive Pulmonary Disease
by Stanislav Kotlyarov
Int. J. Mol. Sci. 2022, 23(2), 985; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23020985 - 17 Jan 2022
Cited by 14 | Viewed by 4084
Abstract
Chronic obstructive pulmonary disease (COPD) is a common, socially significant disease characterized by progressive airflow limitation due to chronic inflammation in the bronchi. Although the causes of COPD are considered to be known, the pathogenesis of the disease continues to be a relevant [...] Read more.
Chronic obstructive pulmonary disease (COPD) is a common, socially significant disease characterized by progressive airflow limitation due to chronic inflammation in the bronchi. Although the causes of COPD are considered to be known, the pathogenesis of the disease continues to be a relevant topic of study. Mechanisms of the innate immune system are involved in various links in the pathogenesis of COPD, leading to persistence of chronic inflammation in the bronchi, their bacterial colonization and disruption of lung structure and function. Bronchial epithelial cells, neutrophils, macrophages and other cells are involved in the development and progression of the disease, demonstrating multiple compromised immune mechanisms. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Host-Pathogen Interaction)
Show Figures

Figure 1

17 pages, 1085 KiB  
Review
The Framework for Human Host Immune Responses to Four Types of Parasitic Infections and Relevant Key JAK/STAT Signaling
by Tsung-Han Wen, Kuo-Wang Tsai, Yan-Jun Wu, Min-Tser Liao, Kuo-Cheng Lu and Wan-Chung Hu
Int. J. Mol. Sci. 2021, 22(24), 13310; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222413310 - 10 Dec 2021
Cited by 12 | Viewed by 4695
Abstract
The human host immune responses to parasitic infections are complex. They can be categorized into four immunological pathways mounted against four types of parasitic infections. For intracellular protozoa, the eradicable host immunological pathway is TH1 immunity involving macrophages (M1), interferon gamma (IFNγ) CD4 [...] Read more.
The human host immune responses to parasitic infections are complex. They can be categorized into four immunological pathways mounted against four types of parasitic infections. For intracellular protozoa, the eradicable host immunological pathway is TH1 immunity involving macrophages (M1), interferon gamma (IFNγ) CD4 T cells, innate lymphoid cells 1 (NKp44+ ILC1), CD8 T cells (Effector-Memory4, EM4), invariant natural killer T cells 1 (iNKT1) cells, and immunoglobulin G3 (IgG3) B cells. For intracellular protozoa, the tolerable host immunological pathway is TH1-like immunity involving macrophages (M2), interferon gamma (IFNγ)/TGFβ CD4 T cells, innate lymphoid cells 1 (NKp44- ILC1), CD8 T cells (EM3), invariant natural killer T 1 (iNKT1) cells, and immunoglobulin A1 (IgA1) B cells. For free-living extracellular protozoa, the eradicable host immunological pathway is TH22 immunity involving neutrophils (N1), interleukin-22 CD4 T cells, innate lymphoid cells 3 (NCR+ ILC3), iNKT17 cells, and IgG2 B cells. For free-living extracellular protozoa, the tolerable host immunological pathway is TH17 immunity involving neutrophils (N2), interleukin-17 CD4 T cells, innate lymphoid cells 3 (NCR- ILC3), iNKT17 cells, and IgA2 B cells. For endoparasites (helminths), the eradicable host immunological pathway is TH2a immunity with inflammatory eosinophils (iEOS), interleukin-5/interleukin-4 CD4 T cells, interleukin-25 induced inflammatory innate lymphoid cells 2 (iILC2), tryptase-positive mast cells (MCt), iNKT2 cells, and IgG4 B cells. For ectoparasites (parasitic insects and arachnids), the eradicable host immunological pathway is TH2b immunity with inflammatory basophils, chymase- and tryptase-positive mast cells (MCct), interleukin-3/interleukin-4 CD4 T cells, interleukin-33 induced nature innate lymphoid cells 2 (nILC2), iNKT2 cells, and immunoglobulin E (IgE) B cells. The tolerable host immunity against ectoparasites and endoparasites is TH9 immunity with regulatory eosinophils, regulatory basophils, interleukin-9 mast cells (MMC9), thymic stromal lymphopoietin induced innate lymphoid cells 2, interleukin-9 CD4 T cells, iNKT2 cells, and IgA2 B cells. In addition, specific transcription factors important for specific immune responses were listed. This JAK/STAT signaling is key to controlling or inducing different immunological pathways. In sum, Tfh is related to STAT5β, and BCL6 expression. Treg is related to STAT5α, STAT5β, and FOXP3. TH1 immunity is related to STAT1α, STAT4, and T-bet. TH2a immunity is related to STAT6, STAT1α, GATA1, and GATA3. TH2b immunity is related to STAT6, STAT3, GATA2, and GATA3. TH22 immunity is associated with both STAT3α and AHR. THαβ immunity is related to STAT1α, STAT1β, STAT2, STAT3β, and ISGF. TH1-like immunity is related to STAT1α, STAT4, STAT5α, and STAT5β. TH9 immunity is related to STAT6, STAT5α, STAT5β, and PU.1. TH17 immunity is related to STAT3α, STAT5α, STAT5β, and RORG. TH3 immunity is related to STAT1α, STAT1β, STAT2, STAT3β, STAT5α, STAT5β, and ISGF. This categorization provides a complete framework of immunological pathways against four types of parasitic infections. This framework as well as relevant JAK/STAT signaling can provide useful knowledge to control allergic hypersensitivities and parasitic infections via development of vaccines or drugs in the near future. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Host-Pathogen Interaction)
Show Figures

Figure 1

14 pages, 1519 KiB  
Review
Pivotal Role of Corneal Fibroblasts in Progression to Corneal Ulcer in Bacterial Keratitis
by Teruo Nishida, Koji Sugioka, Ken Fukuda and Junko Murakami
Int. J. Mol. Sci. 2021, 22(16), 8979; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22168979 - 20 Aug 2021
Cited by 15 | Viewed by 3882
Abstract
The shape and transparency of the cornea are essential for clear vision. However, its location at the ocular surface renders the cornea vulnerable to pathogenic microorganisms in the external environment. Pseudomonas aeruginosa and Staphylococcus aureus are two such microorganisms and are responsible for [...] Read more.
The shape and transparency of the cornea are essential for clear vision. However, its location at the ocular surface renders the cornea vulnerable to pathogenic microorganisms in the external environment. Pseudomonas aeruginosa and Staphylococcus aureus are two such microorganisms and are responsible for most cases of bacterial keratitis. The development of antimicrobial agents has allowed the successful treatment of bacterial keratitis if the infection is diagnosed promptly. However, no effective medical treatment is available after progression to corneal ulcer, which is characterized by excessive degradation of collagen in the corneal stroma and can lead to corneal perforation and corneal blindness. This collagen degradation is mediated by both infecting bacteria and corneal fibroblasts themselves, with a urokinase-type plasminogen activator (uPA)-plasmin-matrix metalloproteinase (MMP) cascade playing a central role in collagen destruction by the host cells. Bacterial factors stimulate the production by corneal fibroblasts of both uPA and pro-MMPs, released uPA mediates the conversion of plasminogen in the extracellular environment to plasmin, and plasmin mediates the conversion of secreted pro-MMPs to the active form of these enzymes, which then degrade stromal collagen. Bacterial factors also stimulate expression by corneal fibroblasts of the chemokine interleukin-8 and the adhesion molecule ICAM-1, both of which contribute to recruitment and activation of polymorphonuclear neutrophils, and these cells then further stimulate corneal fibroblasts via the secretion of interleukin-1. At this stage of the disease, bacteria are no longer necessary for collagen degradation. In this review, we discuss the pivotal role of corneal fibroblasts in corneal ulcer associated with infection by P. aeruginosa or S. aureus as well as the development of potential new modes of treatment for this condition. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Host-Pathogen Interaction)
Show Figures

Figure 1

25 pages, 975 KiB  
Review
Molecular Mechanisms of Lipid Metabolism Disorders in Infectious Exacerbations of Chronic Obstructive Pulmonary Disease
by Stanislav Kotlyarov and Anna Kotlyarova
Int. J. Mol. Sci. 2021, 22(14), 7634; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22147634 - 17 Jul 2021
Cited by 16 | Viewed by 4255
Abstract
Exacerbations largely determine the character of the progression and prognosis of chronic obstructive pulmonary disease (COPD). Exacerbations are connected with changes in the microbiological landscape in the bronchi due to a violation of their immune homeostasis. Many metabolic and immune processes involved in [...] Read more.
Exacerbations largely determine the character of the progression and prognosis of chronic obstructive pulmonary disease (COPD). Exacerbations are connected with changes in the microbiological landscape in the bronchi due to a violation of their immune homeostasis. Many metabolic and immune processes involved in COPD progression are associated with bacterial colonization of the bronchi. The objective of this review is the analysis of the molecular mechanisms of lipid metabolism and immune response disorders in the lungs in COPD exacerbations. The complex role of lipid metabolism disorders in the pathogenesis of some infections is only beginning to be understood, however, there are already fewer and fewer doubts even now about its significance both in the pathogenesis of infectious exacerbations of COPD and in general in the progression of the disease. It is shown that the lipid rafts of the plasma membranes of cells are involved in many processes related to the detection of pathogens, signal transduction, the penetration of pathogens into the cell. Smoking disrupts the normally proceeded processes of lipid metabolism in the lungs, which is a part of the COPD pathogenesis. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Host-Pathogen Interaction)
Show Figures

Figure 1

31 pages, 4593 KiB  
Review
Autophagy, Unfolded Protein Response, and Neuropilin-1 Cross-Talk in SARS-CoV-2 Infection: What Can Be Learned from Other Coronaviruses
by Morvarid Siri, Sanaz Dastghaib, Mozhdeh Zamani, Nasim Rahmani-Kukia, Kiarash Roustai Geraylow, Shima Fakher, Fatemeh Keshvarzi, Parvaneh Mehrbod, Mazaher Ahmadi, Pooneh Mokarram, Kevin M. Coombs and Saeid Ghavami
Int. J. Mol. Sci. 2021, 22(11), 5992; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22115992 - 01 Jun 2021
Cited by 24 | Viewed by 5609
Abstract
The COVID-19 pandemic is caused by the 2019–nCoV/SARS-CoV-2 virus. This severe acute respiratory syndrome is currently a global health emergency and needs much effort to generate an urgent practical treatment to reduce COVID-19 complications and mortality in humans. Viral infection activates various cellular [...] Read more.
The COVID-19 pandemic is caused by the 2019–nCoV/SARS-CoV-2 virus. This severe acute respiratory syndrome is currently a global health emergency and needs much effort to generate an urgent practical treatment to reduce COVID-19 complications and mortality in humans. Viral infection activates various cellular responses in infected cells, including cellular stress responses such as unfolded protein response (UPR) and autophagy, following the inhibition of mTOR. Both UPR and autophagy mechanisms are involved in cellular and tissue homeostasis, apoptosis, innate immunity modulation, and clearance of pathogens such as viral particles. However, during an evolutionary arms race, viruses gain the ability to subvert autophagy and UPR for their benefit. SARS-CoV-2 can enter host cells through binding to cell surface receptors, including angiotensin-converting enzyme 2 (ACE2) and neuropilin-1 (NRP1). ACE2 blockage increases autophagy through mTOR inhibition, leading to gastrointestinal complications during SARS-CoV-2 virus infection. NRP1 is also regulated by the mTOR pathway. An increased NRP1 can enhance the susceptibility of immune system dendritic cells (DCs) to SARS-CoV-2 and induce cytokine storm, which is related to high COVID-19 mortality. Therefore, signaling pathways such as mTOR, UPR, and autophagy may be potential therapeutic targets for COVID-19. Hence, extensive investigations are required to confirm these potentials. Since there is currently no specific treatment for COVID-19 infection, we sought to review and discuss the important roles of autophagy, UPR, and mTOR mechanisms in the regulation of cellular responses to coronavirus infection to help identify new antiviral modalities against SARS-CoV-2 virus. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Host-Pathogen Interaction)
Show Figures

Figure 1

11 pages, 1342 KiB  
Review
AAA+ Molecular Chaperone ClpB in Leptospira interrogans: Its Role and Significance in Leptospiral Virulence and Pathogenesis of Leptospirosis
by Sabina Kędzierska-Mieszkowska and Zbigniew Arent
Int. J. Mol. Sci. 2020, 21(18), 6645; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21186645 - 11 Sep 2020
Cited by 9 | Viewed by 2733
Abstract
Bacterial ClpB is an ATP-dependent disaggregase that belongs to the Hsp100/Clp subfamily of the AAA+ ATPases and cooperates with the DnaK chaperone system in the reactivation of aggregated proteins, as well as promotes bacterial survival under adverse environmental conditions, including thermal and oxidative [...] Read more.
Bacterial ClpB is an ATP-dependent disaggregase that belongs to the Hsp100/Clp subfamily of the AAA+ ATPases and cooperates with the DnaK chaperone system in the reactivation of aggregated proteins, as well as promotes bacterial survival under adverse environmental conditions, including thermal and oxidative stresses. In addition, extensive evidence indicates that ClpB supports the virulence of numerous bacteria, including pathogenic spirochaete Leptospira interrogans responsible for leptospirosis in animals and humans. However, the specific function of ClpB in leptospiral virulence still remains to be fully elucidated. Interestingly, ClpB was predicted as one of the L. interrogans hub proteins interacting with human proteins, and pathogen–host protein interactions are fundamental for successful invasion of the host immune system by bacteria. The aim of this review is to discuss the most important aspects of ClpB’s function in L. interrogans, including contribution of ClpB to leptospiral virulence and pathogenesis of leptospirosis, a zoonotic disease with a significant impact on public health worldwide. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Host-Pathogen Interaction)
Show Figures

Figure 1

34 pages, 4948 KiB  
Review
Adherent-Invasive E. coli: Update on the Lifestyle of a Troublemaker in Crohn’s Disease
by Mélissa Chervy, Nicolas Barnich and Jérémy Denizot
Int. J. Mol. Sci. 2020, 21(10), 3734; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21103734 - 25 May 2020
Cited by 58 | Viewed by 6741
Abstract
Besides genetic polymorphisms and environmental factors, the intestinal microbiota is an important factor in the etiology of Crohn’s disease (CD). Among microbiota alterations, a particular pathotype of Escherichia coli involved in the pathogenesis of CD abnormally colonizes the intestinal mucosa of patients: the [...] Read more.
Besides genetic polymorphisms and environmental factors, the intestinal microbiota is an important factor in the etiology of Crohn’s disease (CD). Among microbiota alterations, a particular pathotype of Escherichia coli involved in the pathogenesis of CD abnormally colonizes the intestinal mucosa of patients: the adherent-invasive Escherichia coli (AIEC) pathobiont bacteria, which have the abilities to adhere to and to invade intestinal epithelial cells (IECs), as well as to survive and replicate within macrophages. AIEC have been the subject of many studies in recent years to unveil some genes linked to AIEC virulence and to understand the impact of AIEC infection on the gut and consequently their involvement in CD. In this review, we describe the lifestyle of AIEC bacteria within the intestine, from the interaction with intestinal epithelial and immune cells with an emphasis on environmental and genetic factors favoring their implantation, to their lifestyle in the intestinal lumen. Finally, we discuss AIEC-targeting strategies such as the use of FimH antagonists, bacteriophages, or antibiotics, which could constitute therapeutic options to prevent and limit AIEC colonization in CD patients. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Host-Pathogen Interaction)
Show Figures

Figure 1

Other

Jump to: Research, Review

10 pages, 2151 KiB  
Brief Report
Staphylococcus aureus Decreases SUMOylation Host Response to Promote Intramacrophage Survival
by Nadhuma Youssouf, Clara Recasens-Zorzo, Virginie Molle, Guillaume Bossis, Philippe Soubeyran and Laila Gannoun-Zaki
Int. J. Mol. Sci. 2021, 22(15), 8108; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22158108 - 28 Jul 2021
Cited by 6 | Viewed by 2121
Abstract
Staphylococcus aureus is a commensal bacterium that causes severe infections in soft tissue and the bloodstream. During infection, S. aureus manipulates host cell response to facilitate its own replication and dissemination. Here, we show that S. aureus significantly decreases the level of SUMOylation, [...] Read more.
Staphylococcus aureus is a commensal bacterium that causes severe infections in soft tissue and the bloodstream. During infection, S. aureus manipulates host cell response to facilitate its own replication and dissemination. Here, we show that S. aureus significantly decreases the level of SUMOylation, an essential post-translational modification, in infected macrophages 24 h post-phagocytosis. The reduced level of SUMOylation correlates with a decrease in the SUMO-conjugating enzyme Ubc9. The over-expression of SUMO proteins in macrophages impaired bacterial intracellular proliferation and the inhibition of SUMOylation with ML-792 increased it. Together, these findings demonstrated for the first time the role of host SUMOylation response toward S. aureus infection. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Host-Pathogen Interaction)
Show Figures

Figure 1

Back to TopTop