Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 507
Filter
2.
Sci Immunol ; 6(65): eabj2132, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34797692

ABSTRACT

Alveolar macrophages are the most abundant macrophages in the healthy lung where they play key roles in homeostasis and immune surveillance against airborne pathogens. Tissue-specific differentiation and survival of alveolar macrophages rely on niche-derived factors, such as granulocyte-macrophage colony-stimulating factor (GM-CSF) and transforming growth factor­ß (TGF-ß). However, the nature of the downstream molecular pathways that regulate the identity and function of alveolar macrophages and their response to injury remain poorly understood. Here, we identify that the transcription factor EGR2 is an evolutionarily conserved feature of lung alveolar macrophages and show that cell-intrinsic EGR2 is indispensable for the tissue-specific identity of alveolar macrophages. Mechanistically, we show that EGR2 is driven by TGF-ß and GM-CSF in a PPAR-γ­dependent manner to control alveolar macrophage differentiation. Functionally, EGR2 was dispensable for the regulation of lipids in the airways but crucial for the effective handling of the respiratory pathogen Streptococcus pneumoniae. Last, we show that EGR2 is required for repopulation of the alveolar niche after sterile, bleomycin-induced lung injury and demonstrate that EGR2-dependent, monocyte-derived alveolar macrophages are vital for effective tissue repair after injury. Collectively, we demonstrate that EGR2 is an indispensable component of the transcriptional network controlling the identity and function of alveolar macrophages in health and disease.


Subject(s)
Early Growth Response Protein 2/immunology , Macrophages, Alveolar/immunology , Animals , Female , Humans , Macrophages, Alveolar/pathology , Male , Mice , Pneumococcal Infections/immunology , Pneumococcal Infections/pathology , Streptococcus pneumoniae/immunology
3.
J Cell Mol Med ; 25(16): 7621-7630, 2021 08.
Article in English | MEDLINE | ID: mdl-34272809

ABSTRACT

Community-acquired pneumonia (CAP) is a major cause of sepsis. Despite several clinical trials targeting components of the inflammatory response, no specific treatment other than antimicrobial therapy has been approved. This argued for a deeper understanding of sepsis immunopathology, in particular factors that can modulate the host response. Small non-coding RNA, for example, micro (mi)RNA, have been established as important modifiers of cellular phenotypes. Notably, miRNAs are not exclusive to the intracellular milieu but have also been detected extracellular in the circulation with functional consequences. Here, we sought to determine shifts in circulatory small RNA levels of critically ill patients with CAP-associated sepsis and to determine the influence of clinical severity and causal pathogens on small RNA levels. Blood plasma was collected from 13 critically ill patients with sepsis caused by CAP on intensive care unit admission and from 5 non-infectious control participants. Plasma small RNA-sequencing identified significantly altered levels of primarily mature miRNAs in CAP relative to controls. Pathways analysis of high or low abundance miRNA identified various over-represented cellular biological pathways. Analysis of small RNA levels against common clinical severity and inflammatory parameters indices showed direct and indirect correlations. Additionally, variance of plasma small RNA levels in CAP patients may be explained, at least in part, by differences in causal pathogens. Small nuclear RNA levels were specifically altered in CAP due to Influenza infection in contrast to Streptococcus pneumoniae infection. Pathway analysis of plasma miRNA signatures unique to Influenza or Streptococcus pneumoniae infections showed enrichment for specific proteoglycan, cell cycle, and immunometabolic pathways.


Subject(s)
Community-Acquired Infections/pathology , MicroRNAs/genetics , Pneumococcal Infections/pathology , Pneumonia/pathology , RNA, Small Untranslated/genetics , Sepsis/pathology , Streptococcus pneumoniae/genetics , Aged , Community-Acquired Infections/blood , Community-Acquired Infections/genetics , Community-Acquired Infections/microbiology , Female , Humans , Intensive Care Units/organization & administration , Male , MicroRNAs/blood , Middle Aged , Pneumococcal Infections/blood , Pneumococcal Infections/genetics , Pneumococcal Infections/microbiology , Pneumonia/blood , Pneumonia/genetics , Pneumonia/microbiology , RNA, Small Untranslated/blood , Sepsis/blood , Sepsis/genetics , Severity of Illness Index , Streptococcus pneumoniae/isolation & purification , Streptococcus pneumoniae/pathogenicity
4.
Brain Res Bull ; 174: 296-304, 2021 09.
Article in English | MEDLINE | ID: mdl-34216650

ABSTRACT

Cerebral ischaemia is accompanied by infectious complications due to immunosuppression, known as stroke-induced immunodepression (SIID). Orexin-A (OXA), a neuropeptide produced in the hypothalamus, has been reported to have neuroprotective properties after stroke and is known to modulate inflammatory processes in peripheral tissues. The aim of this study was to determine the effects of orexin-A (OXA) on cerebral ischaemic inflammatory injury and SIID following experimental stroke. Cerebral ischaemia was induced in C57/BL6 mice by middle cerebral artery occlusion (MCAO). A mouse model of pneumonia and poststroke pneumococcal pneumonia was established by intratracheal inoculation with S. pneumoniae in a normal mouse or MCAO mouse model on the third day. We found that OXA postconditioning inhibited cerebral ischaemic inflammatory injury. The mechanism involved downregulation of the NF-κB signalling pathway. In addition, OXA may serve as a potential treatment target for attenuating stroke-induced immunodepression in mice.


Subject(s)
Brain Ischemia/drug therapy , Encephalitis/drug therapy , NF-kappa B/drug effects , Neuroprotective Agents/therapeutic use , Signal Transduction/drug effects , Stroke/immunology , Transcription Factor RelA/therapeutic use , Animals , Immune Tolerance/drug effects , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , Male , Mice , Mice, Inbred C57BL , Pneumococcal Infections/complications , Pneumococcal Infections/drug therapy , Pneumococcal Infections/pathology , Stroke/drug therapy
5.
J Biol Chem ; 297(2): 101000, 2021 08.
Article in English | MEDLINE | ID: mdl-34303706

ABSTRACT

DNA gyrase is a type II topoisomerase that is responsible for maintaining the topological state of bacterial and some archaeal genomes. It uses an ATP-dependent two-gate strand-passage mechanism that is shared among all type II topoisomerases. During this process, DNA gyrase creates a transient break in the DNA, the G-segment, to form a cleavage complex. This allows a second DNA duplex, known as the T-segment, to pass through the broken G-segment. After the broken strand is religated, the T-segment is able to exit out of the enzyme through a gate called the C-gate. Although many steps of the type II topoisomerase mechanism have been studied extensively, many questions remain about how the T-segment ultimately exits out of the C-gate. A recent cryo-EM structure of Streptococcus pneumoniae GyrA shows a putative T-segment in close proximity to the C-gate, suggesting that residues in this region may be important for coordinating DNA exit from the enzyme. Here, we show through site-directed mutagenesis and biochemical characterization that three conserved basic residues in the C-gate of DNA gyrase are important for DNA supercoiling activity, but not for ATPase or cleavage activity. Together with the structural information previously published, our data suggest a model in which these residues cluster to form a positively charged region that facilitates T-segment passage into the cavity formed between the DNA gate and C-gate.


Subject(s)
Catalytic Domain , DNA Gyrase/metabolism , DNA, Bacterial/chemistry , DNA, Superhelical , Pneumococcal Infections/enzymology , Protein Structural Elements , Streptococcus pneumoniae/enzymology , DNA Gyrase/chemistry , DNA Topoisomerases, Type II/chemistry , DNA Topoisomerases, Type II/metabolism , Models, Molecular , Mutagenesis, Site-Directed/methods , Pneumococcal Infections/microbiology , Pneumococcal Infections/pathology , Streptococcus pneumoniae/isolation & purification , Streptococcus pneumoniae/pathogenicity
6.
Front Immunol ; 12: 624775, 2021.
Article in English | MEDLINE | ID: mdl-33953708

ABSTRACT

Objective: Streptococcus pneumoniae (S.pn) is a common respiratory pathogen and a frequent cause of acute otitis media (AOM) in children. However, little is known about the immunometabolism during AOM. This study was to assess the presence of glucose metabolic reprogramming during AOM and its underlying mechanism affecting inflammatory response and middle ear injury. Methods: The levels of glycolytic metabolism were evaluated by measuring the expression of glycolysis-related genes and the production of metabolites. HE stain, immunofluorescence, immunohistochemistry, enzyme-linked immunosorbent assay (ELISA) and Western blot were performed to measure the effect of glucose metabolic reprogramming on inflammatory response, pneumococcal clearance, hypoxia-inducible factor 1 alpha (HIF-1α) expression and cytokine secretion during AOM, respectively. Results: The analysis of microarray revealed an increase of the expression of glycolysis-related genes during S.pn-induced AOM, which was verified by real-time PCR. Increased glycolysis promoted the production of IL-1ß and TNF-α and facilitated the clearance of S.pn by enhancing phagocytosis and killing capability of neutrophils, but also aggravated the middle ear injury. Furthermore, these pathogenic effects could be reversed after glycolytic inhibitor 2DG treatment. Additionally, HIF-1α was observed to involve in glycolytic metabolism during AOM. Conclusion: S.pn infection induced increased glycolysis conversion during AOM, which promoted inflammatory responses and bacterial clearance, but also aggravated tissue damage.


Subject(s)
Ear, Middle/metabolism , Glycolysis , Otitis Media/metabolism , Pneumococcal Infections/metabolism , Streptococcus pneumoniae/pathogenicity , Animals , Disease Models, Animal , Ear, Middle/immunology , Ear, Middle/microbiology , Ear, Middle/pathology , Gene Expression Regulation, Enzymologic , Host-Pathogen Interactions , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Mice, Inbred C57BL , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/microbiology , Otitis Media/immunology , Otitis Media/microbiology , Otitis Media/pathology , Phagocytosis , Pneumococcal Infections/immunology , Pneumococcal Infections/microbiology , Pneumococcal Infections/pathology , Streptococcus pneumoniae/immunology , Tumor Necrosis Factor-alpha/metabolism
7.
Front Cell Infect Microbiol ; 11: 643326, 2021.
Article in English | MEDLINE | ID: mdl-33828999

ABSTRACT

Secondary bacterial infections enhance the disease burden of influenza infections substantially. Streptococcus pneumoniae (the pneumococcus) plays a major role in the synergism between bacterial and viral pathogens, which is based on complex interactions between the pathogen and the host immune response. Here, we discuss mechanisms that drive the pathogenesis of a secondary pneumococcal infection after an influenza infection with a focus on how pneumococci senses and adapts to the influenza-modified environment. We briefly summarize what is known regarding secondary bacterial infection in relation to COVID-19 and highlight the need to improve our current strategies to prevent and treat viral bacterial coinfections.


Subject(s)
COVID-19/complications , Influenza, Human , Pneumococcal Infections/pathology , Respiratory System/pathology , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/virology , Coinfection , Host-Pathogen Interactions/immunology , Humans , Influenza, Human/complications , Streptococcus pneumoniae
8.
Int J Mol Sci ; 22(8)2021 Apr 08.
Article in English | MEDLINE | ID: mdl-33918100

ABSTRACT

Streptococcus pneumoniae (S. pneumoniae) causes severe pulmonary diseases, leading to high morbidity and mortality. It has been reported that inflammasomes such as NLR family pyrin domain containing 3 (NLRP3) and absent in melanoma 2 (AIM2) play an important role in the host defense against S. pneumoniae infection. However, the role of NLRP6 in vivo and in vitro against S. pneumoniae remains unclear. Therefore, we investigated the role of NLRP6 in regulating the S. pneumoniae-induced inflammatory signaling pathway in vitro and the role of NLRP6 in the host defense against S. pneumoniae in vivo by using NLRP6-/- mice. The results showed that the NLRP6 inflammasome regulated the maturation and secretion of IL-1ß, but it did not affect the induction of IL-1ß transcription in S. pneumoniae-infected macrophages. Furthermore, the activation of caspase-1, caspase-11, and gasdermin D (GSDMD) as well as the oligomerization of apoptosis-associated speck-like protein (ASC) were also mediated by NLRP6 in S. pneumoniae-infected macrophages. However, the activation of NLRP6 reduced the expression of NF-κB and ERK signaling pathways in S. pneumoniae-infected macrophages. In vivo study showed that NLRP6-/- mice had a higher survival rate, lower number of bacteria, and milder inflammatory response in the lung compared with wild-type (WT) mice during S. pneumoniae infection, indicating that NLRP6 plays a negative role in the host defense against S. pneumoniae. Furthermore, increased bacterial clearance in NLRP6 deficient mice was modulated by the recruitment of macrophages and neutrophils. Our study provides a new insight on S. pneumoniae-induced activation of NLRP6 and suggests that blocking NLRP6 could be considered as a potential therapeutic strategy to treat S. pneumoniae infection.


Subject(s)
Host-Pathogen Interactions , Inflammasomes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Pneumococcal Infections/metabolism , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/physiology , Animals , Caspase 1/metabolism , Caspases, Initiator/metabolism , Cytokines/biosynthesis , Disease Models, Animal , Disease Susceptibility , Intracellular Signaling Peptides and Proteins/genetics , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Pneumococcal Infections/immunology , Pneumococcal Infections/pathology , Signal Transduction
9.
Infect Immun ; 89(7): e0002321, 2021 06 16.
Article in English | MEDLINE | ID: mdl-33875471

ABSTRACT

Streptococcus pneumoniae (pneumococcus) is one of the primary bacterial pathogens that complicates influenza virus infections. These bacterial coinfections increase influenza-associated morbidity and mortality through a number of immunological and viral-mediated mechanisms, but the specific bacterial genes that contribute to postinfluenza pathogenicity are not known. Here, we used genome-wide transposon mutagenesis (Tn-Seq) to reveal bacterial genes that confer improved fitness in influenza virus-infected hosts. The majority of the 32 genes identified are involved in bacterial metabolism, including nucleotide biosynthesis, amino acid biosynthesis, protein translation, and membrane transport. We generated mutants with single-gene deletions (SGD) of five of the genes identified, SPD1414, SPD2047 (cbiO1), SPD0058 (purD), SPD1098, and SPD0822 (proB), to investigate their effects on in vivo fitness, disease severity, and host immune responses. The growth of the SGD mutants was slightly attenuated in vitro and in vivo, but each still grew to high titers in the lungs of mock- and influenza virus-infected hosts. Despite high bacterial loads, mortality was significantly reduced or delayed with all SGD mutants. Time-dependent reductions in pulmonary neutrophils, inflammatory macrophages, and select proinflammatory cytokines and chemokines were also observed. Immunohistochemical staining further revealed altered neutrophil distribution with reduced degeneration in the lungs of influenza virus-SGD mutant-coinfected animals. These studies demonstrate a critical role for specific bacterial genes and for bacterial metabolism in driving virulence and modulating immune function during influenza-associated bacterial pneumonia.


Subject(s)
Coinfection , Genetic Fitness , Host-Pathogen Interactions , Influenza A virus , Influenza, Human/virology , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/physiology , Bacterial Proteins/genetics , Cytokines/metabolism , Host-Pathogen Interactions/immunology , Humans , Inflammation Mediators , Influenza A virus/immunology , Leukocytes/immunology , Leukocytes/metabolism , Mutation , Pneumococcal Infections/immunology , Pneumococcal Infections/pathology
11.
Mol Microbiol ; 116(2): 438-458, 2021 08.
Article in English | MEDLINE | ID: mdl-33811693

ABSTRACT

Streptococcus pneumoniae resides in the human upper airway as a commensal but also causes pneumonia, bacteremia, meningitis, and otitis media. It remains unclear how pneumococci adapt to nutritional conditions of various host niches. We here show that MetR, a LysR family transcriptional regulator, serves as a molecular adaptor for pneumococcal fitness, particularly in the upper airway. The metR mutant of strain D39 rapidly disappeared from the nasopharynx but was marginally attenuated in the lungs and bloodstream of mice. RNA-seq and ChIP-seq analyses showed that MetR broadly regulates transcription of the genes involved in methionine synthesis and other functions under methionine starvation. Genetic and biochemical analyses confirmed that MetR is essential for the activation of methionine synthesis but not uptake. Co-infection of influenza virus partially restored the colonization defect of the metR mutant. These results strongly suggest that MetR is particularly evolved for pneumococcal carriage in the upper airway of healthy individuals where free methionine is severely limited, but it becomes dispensable where environmental methionine is relatively more abundant (e.g., inflamed upper airway and sterile sites). To the best of our knowledge, MetR represents the first known regulator particularly for pneumococcal carriage in healthy individuals.


Subject(s)
Bacterial Proteins/genetics , Methionine/biosynthesis , Nasopharynx/microbiology , Streptococcus pneumoniae/growth & development , Streptococcus pneumoniae/genetics , Trans-Activators/genetics , Animals , Bacterial Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Methionine/metabolism , Mice , Pneumococcal Infections/pathology , Trans-Activators/metabolism , Transcription, Genetic/genetics
12.
PLoS One ; 16(3): e0235026, 2021.
Article in English | MEDLINE | ID: mdl-33705390

ABSTRACT

Respiratory syncytial virus (RSV) is the primary cause of viral bronchiolitis resulting in hospitalization and a frequent cause of secondary respiratory bacterial infection, especially by Streptococcus pneumoniae (Spn) in infants. While murine studies have demonstrated enhanced morbidity during a viral/bacterial co-infection, human meta-studies have conflicting results. Moreover, little knowledge about the pathogenesis of emerging Spn serotype 22F, especially the co-pathologies between RSV and Spn, is known. Here, colostrum-deprived neonate lambs were divided into four groups. Two of the groups were nebulized with RSV M37, and the other two groups were mock nebulized. At day three post-RSV infection, one RSV group (RSV/Spn) and one mock-nebulized group (Spn only) were inoculated with Spn intratracheally. At day six post-RSV infection, bacterial/viral loads were assessed along with histopathology and correlated with clinical symptoms. Lambs dually infected with RSV/Spn trended with higher RSV titers, but lower Spn. Additionally, lung lesions were observed to be more frequent in the RSV/Spn group characterized by increased interalveolar wall thickness accompanied by neutrophil and lymphocyte infiltration and higher myeloperoxidase. Despite lower Spn in lungs, co-infected lambs had more significant morbidity and histopathology, which correlated with a different cytokine response. Thus, enhanced disease severity during dual infection may be due to lesion development and altered immune responses rather than bacterial counts.


Subject(s)
Pneumococcal Infections/pathology , Respiratory Syncytial Virus Infections/pathology , Streptococcus pneumoniae/isolation & purification , Animals , Animals, Newborn , Bronchoalveolar Lavage Fluid/microbiology , Bronchoalveolar Lavage Fluid/virology , Cytokines/metabolism , Disease Models, Animal , Lung/microbiology , Lung/pathology , Lung/virology , Lymphocytes/cytology , Lymphocytes/immunology , Neutrophils/cytology , Neutrophils/immunology , Peroxidase/metabolism , Pneumococcal Infections/complications , Pneumococcal Infections/epidemiology , Pneumococcal Infections/microbiology , RNA, Viral/metabolism , Respiratory Syncytial Virus Infections/complications , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Viruses/genetics , Respiratory Syncytial Viruses/isolation & purification , Serogroup , Sheep , Streptococcus pneumoniae/genetics
13.
Sci Rep ; 11(1): 6195, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33737573

ABSTRACT

Streptococcus pneumoniae endophthalmitis is clinically more severe, more difficult to treat, and carry a higher risk of vision loss, evisceration, or enucleation. This study is to investigate the clinical settings, antibiotic susceptibility, and visual outcomes of S. pneumoniae endophthalmitis at a tertiary referral center in Taiwan. S. pneumoniae endophthalmitis was diagnosed in 38 eyes of 38 patients. The main clinical features were postcataract endophthalmitis (n = 13, 34%) and endophthalmitis associated with corneal ulcer (n = 12, 32%), trauma (n = 6, 16%), endogenous etiology (n = 4, 11%), trabeculectomy (n = 2, 5%), and pterygium excision-related scleral ulcer (n = 1, 3%). Presenting visual acuity ranged from counting fingers to no light perception. Pars plana vitrectomy with intravitreal antibiotics was performed in 17 eyes (39%) in primary or secondary treatments. S. pneumoniae isolates were susceptible to vancomycin (38/38, 100%), penicillin (37/38, 97%), ceftriaxone (37/38, 97%), cefuroxime (12/15, 80%), levofloxacin (13/15 ,87%), and moxifloxacin (15/17, 88%). Final visual acuity was better than 20/400 in 3 of 38 eyes (8%), 5/200 to hand motions in 3 eyes (8%), and light perception to no light perception in 32 eyes (84%). Ten eyes (26%) underwent evisceration or enucleation. Although S. pneumoniae isolates were susceptible to vancomycin, S. pneumoniae endophthalmitis had a very poor visual prognosis.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Endophthalmitis/pathology , Pneumococcal Infections/pathology , Streptococcus pneumoniae/pathogenicity , Vitrectomy/statistics & numerical data , Adult , Aged , Aged, 80 and over , Cataract/complications , Cataract/microbiology , Cataract/pathology , Cataract Extraction/adverse effects , Ceftriaxone/therapeutic use , Cefuroxime/therapeutic use , Corneal Ulcer/complications , Corneal Ulcer/microbiology , Corneal Ulcer/pathology , Endophthalmitis/etiology , Endophthalmitis/microbiology , Eye Enucleation/methods , Eye Enucleation/statistics & numerical data , Eye Injuries/complications , Eye Injuries/microbiology , Eye Injuries/pathology , Female , Humans , Levofloxacin/therapeutic use , Male , Microbial Sensitivity Tests , Middle Aged , Moxifloxacin/therapeutic use , Penicillins/therapeutic use , Pneumococcal Infections/etiology , Pneumococcal Infections/microbiology , Retrospective Studies , Severity of Illness Index , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/growth & development , Taiwan , Tertiary Care Centers , Trabeculectomy/adverse effects , Treatment Outcome , Vancomycin/therapeutic use , Vitrectomy/methods
14.
Sci Rep ; 11(1): 1701, 2021 01 18.
Article in English | MEDLINE | ID: mdl-33462258

ABSTRACT

The PspC and Hic proteins of Streptococcus pneumoniae are some of the most variable microbial immune evasion proteins identified to date. Due to structural similarities and conserved binding profiles, it was assumed for a long time that these pneumococcal surface proteins represent a protein family comprised of eleven subgroups. Recently, however, the evaluation of more proteins revealed a greater diversity of individual proteins. In contrast to previous assumptions a pattern evaluation of six PspC and five Hic variants, each representing one of the previously defined subgroups, revealed distinct structural and likely functionally regions of the proteins, and identified nine new domains and new domain alternates. Several domains are unique to PspC and Hic variants, while other domains are also present in other virulence factors encoded by pneumococci and other bacterial pathogens. This knowledge improved pattern evaluation at the level of full-length proteins, allowed a sequence comparison at the domain level and identified domains with a modular composition. This novel strategy increased understanding of individual proteins variability and modular domain composition, enabled a structural and functional characterization at the domain level and furthermore revealed substantial structural differences between PspC and Hic proteins. Given the exceptional genomic diversity of the multifunctional PspC and Hic proteins a detailed structural and functional evaluation need to be performed at the strain level. Such knowledge will also be useful for molecular strain typing and characterizing PspC and Hic proteins from new clinical S. pneumoniae strains.


Subject(s)
Bacterial Proteins/chemistry , Carrier Proteins/chemistry , Streptococcus pneumoniae/metabolism , Bacterial Proteins/classification , Bacterial Proteins/genetics , Carrier Proteins/classification , Carrier Proteins/genetics , Humans , Mutation , Phylogeny , Pneumococcal Infections/immunology , Pneumococcal Infections/microbiology , Pneumococcal Infections/pathology , Protein Binding , Protein Domains , Sequence Analysis, Protein , Streptococcus pneumoniae/isolation & purification
16.
Emerg Infect Dis ; 27(3): 919-923, 2021 03.
Article in English | MEDLINE | ID: mdl-33443011

ABSTRACT

Postmortem lung pathology of a patient in Japan with severe acute respiratory syndrome coronavirus 2 infection showed diffuse alveolar damage as well as bronchopneumonia caused by Streptococcus pneumoniae infection. The distribution of each pathogen and the accompanying histopathology suggested the infections progressed in a mutually exclusive manner within the lung, resulting in fatal respiratory failure.


Subject(s)
COVID-19/pathology , Coinfection , Lung , Pneumococcal Infections/pathology , Aged, 80 and over , Autopsy , Humans , Lung/microbiology , Lung/pathology , Lung/virology , Male , SARS-CoV-2/isolation & purification , Streptococcus pneumoniae/isolation & purification
17.
Pharmacol Res ; 163: 105292, 2021 01.
Article in English | MEDLINE | ID: mdl-33171305

ABSTRACT

Resolution failure of exacerbated inflammation triggered by Influenza A virus (IAV) prevents return of pulmonary homeostasis and survival, especially when associated with secondary pneumococcal infection. Therapeutic strategies based on pro-resolving molecules have great potential against acute inflammatory diseases. Angiotensin-(1-7) [Ang-(1-7)] is a pro-resolving mediator that acts on its Mas receptor (MasR) to promote resolution of inflammation. We investigated the effects of Ang-(1-7) and the role of MasR in the context of primary IAV infection and secondary pneumococcal infection and evaluated pulmonary inflammation, virus titers and bacteria counts, and pulmonary damage. Therapeutic treatment with Ang-(1-7) decreased neutrophil recruitment, lung injury, viral load and morbidity after a primary IAV infection. Ang-(1-7) induced apoptosis of neutrophils and efferocytosis of these cells by alveolar macrophages, but had no direct effect on IAV replication in vitro. MasR-deficient (MasR-/-) mice were highly susceptible to IAV infection, displaying uncontrolled inflammation, increased viral load and greater lethality rate, as compared to WT animals. Ang-(1-7) was not protective in MasR-/- mice. Interestingly, Ang-(1-7) given during a sublethal dose of IAV infection greatly reduced morbidity associated with a subsequent S. pneumoniae infection, as seen by decrease in the magnitude of neutrophil influx, number of bacteria in the blood leading to a lower lethality. Altogether, these results show that Ang-(1-7) is highly protective against severe primary IAV infection and protects against secondary bacterial infection of the lung. These effects are MasR-dependent. Mediators of resolution of inflammation, such as Ang-(1-7), should be considered for the treatment of pulmonary viral infections.


Subject(s)
Angiotensin I/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Peptide Fragments/therapeutic use , Pneumococcal Infections/drug therapy , Pneumonia, Viral/drug therapy , Proto-Oncogene Proteins/immunology , Receptors, G-Protein-Coupled/immunology , A549 Cells , Angiotensin I/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cytokines/immunology , Dogs , Humans , Influenza A virus , Lung/drug effects , Lung/immunology , Lung/pathology , Madin Darby Canine Kidney Cells , Male , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/drug effects , Neutrophils/immunology , Peptide Fragments/pharmacology , Peroxidase/immunology , Phagocytosis/drug effects , Pneumococcal Infections/immunology , Pneumococcal Infections/pathology , Pneumonia, Viral/immunology , Pneumonia, Viral/pathology , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Streptococcus pneumoniae
18.
Ann Emerg Med ; 77(1): 11-18, 2021 01.
Article in English | MEDLINE | ID: mdl-32747082

ABSTRACT

STUDY OBJECTIVE: This study sought to describe the clinical presentation of normocellular community-acquired bacterial meningitis in adults. METHODS: Using the prospective, nationwide, population-based database of the Danish Study Group of Infections of the Brain, the study identified all adults with normocellular community-acquired bacterial meningitis who were treated at departments of infectious diseases in Denmark from 2015 through 2018. Normocellular community-acquired bacterial meningitis was defined as a cerebrospinal fluid leukocyte count of up to 10×106/L combined with detection of bacteria in the cerebrospinal fluid. Outcome was categorized according to the Glasgow Outcome Scale at discharge. RESULTS: Normocellular cerebrospinal fluid was observed in 12 of 696 (2%) patients with community-acquired bacterial meningitis. The median age was 70 years (range 17 to 92 years), and 8 of 12 (67%) patients were male. All patients had symptoms suggestive of community-acquired bacterial meningitis and pathogens identified by culture (Streptococcus pneumoniae, n=10; Staphylococcus aureus, n=1) or polymerase chain reaction (Neisseria meningitidis; n=1) of the cerebrospinal fluid. Bacteremia was found in 9 of 12 (75%) patients, and 1 of 12 (8%) presented with septic shock. None of the patients had serious underlying immunocompromising conditions. The median times from admission to lumbar puncture and meningitis treatment were 2.5 hours (interquartile range 1.1 to 3.9 hours) and 2.6 hours (interquartile range 0.9 to 22.8 hours). In 3 of 11 (27%) patients, empiric treatment for community-acquired bacterial meningitis was interrupted by a normal cerebrospinal fluid cell count. The overall case-fatality rate was 3 of 12 (25%); meningitis treatment was interrupted in 1 of these patients, and 8 of 12 (67%) had a Glasgow Outcome Scale score of 1 to 4 at discharge. CONCLUSION: Normocellular community-acquired bacterial meningitis is not very common, but it is important to consider and may be associated with a pneumococcal cause.


Subject(s)
Meningitis, Bacterial/diagnosis , Adolescent , Adult , Aged , Aged, 80 and over , Community-Acquired Infections , Denmark , Female , Humans , Leukocyte Count , Male , Meningitis, Bacterial/cerebrospinal fluid , Meningitis, Bacterial/microbiology , Meningitis, Bacterial/pathology , Meningococcal Infections/cerebrospinal fluid , Meningococcal Infections/diagnosis , Meningococcal Infections/microbiology , Meningococcal Infections/pathology , Middle Aged , Neisseria meningitidis , Pneumococcal Infections/cerebrospinal fluid , Pneumococcal Infections/diagnosis , Pneumococcal Infections/microbiology , Pneumococcal Infections/pathology , Prospective Studies , Staphylococcal Infections/cerebrospinal fluid , Staphylococcal Infections/diagnosis , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Streptococcus pneumoniae , Young Adult
19.
BMC Microbiol ; 20(1): 376, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33334315

ABSTRACT

BACKGROUND: Streptococcus pneumonia (pneumococcus) is a human bacterial pathogen causing a range of mild to severe infections. The complicated transcriptome patterns of pneumococci during the colonization to infection process in the human body are usually determined by measuring the expression of essential virulence genes and the comparison of pathogenic with non-pathogenic bacteria through microarray analyses. As systems biology studies have demonstrated, critical co-expressing modules and genes may serve as key players in biological processes. Generally, Sample Progression Discovery (SPD) is a computational approach traditionally used to decipher biological progression trends and their corresponding gene modules (clusters) in different clinical samples underlying a microarray dataset. The present study aimed to investigate the bacterial gene expression pattern from colonization to severe infection periods (specimens isolated from the nasopharynx, lung, blood, and brain) to find new genes/gene modules associated with the infection progression. This strategy may lead to finding novel gene candidates for vaccines or drug design. RESULTS: The results included essential genes whose expression patterns varied in different bacterial conditions and have not been investigated in similar studies. CONCLUSIONS: In conclusion, the SPD algorithm, along with differentially expressed genes detection, can offer new ways of discovering new therapeutic or vaccine targeted gene products.


Subject(s)
Gene Regulatory Networks , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/pathogenicity , Algorithms , Animals , Disease Progression , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Genes, Bacterial/genetics , Mice , Pneumococcal Infections/microbiology , Pneumococcal Infections/pathology , Systems Biology , Virulence/genetics
20.
Med. clín (Ed. impr.) ; 155(11): 502-505, dic. 2020. tab, ilus
Article in English | IBECS | ID: ibc-190157

ABSTRACT

BACKGROUND: In the context of the COVID-19 pandemic the risk of misdiagnosis of other causes of respiratory infection is likely. In this work we aim to describe the clinical characteristics, treatment and outcome of pneumococcal infection in COVID-19 patients. PATIENTS AND METHODS: Every COVID-19 patient presenting with concomitant pneumococcal pneumonia during March 2020 in a tertiary teaching Hospital In Barcelona, Spain. RESULTS: Five patients with PCR confirmed COVID19 or clinical and radiological suspicion were diagnosed of pneumococcal infection. In all cases chest X-ray were abnormal, with unilateral or bilateral infiltrates. Procalcitonin showed to be not sensitive enough to detect pneumococcal infection. Antibiotherapy was promptly started in all five cases with subsequent satisfactory evolution. CONCLUSION: International guidelines do not include the universal screening for bacterial co-infection. Radiological pattern of COVID-19 can be indistinguishable from that of pneumococcus pneumonia and frequency of co-infection is not well stablished, therefore clinicians should be aware of the possible SARS-CoV-2-pneumococcus association to avoid misdiagnosis and delay antibiotic therapy


INTRODUCCIÓN: En el contexto de la pandemia por COVID-19 el riesgo de errores en el diagnóstico de otras causas de infección respiratoria es elevado. En este trabajo describimos las características clínicas, el tratamiento y la evolución de los pacientes con coinfección por COVID-19 y neumococo. PACIENTES Y MÉTODOS: Todos los pacientes con COVID-19 que presentaron neumonía neumocócica durante marzo 2020 en un hospital universitario de Barcelona, España. RESULTADOS: Cinco pacientes con COVID-19 confirmada por PCR o sospecha radiológica fueron diagnosticados de infección por neumococo. En todos los casos la radiografía de tórax era patológica con infiltrado unilateral o bilateral. La procalcitonina demostró no ser suficientemente sensible para detectar la infección neumocócica. La antibioterapia fue iniciada de manera precoz en los 5 casos con evolución satisfactoria. CONCLUSIONES: Las guías internacionales no incluyen el cribado universal para coinfección bacteriana. El patrón radiológico del COVID-19 puede ser indistinguible de la neumonía neumocócica, y la frecuencia de la coinfección no ha sido establecida. Los clínicos deben de ser conscientes de la posible asociación de SARS-CoV-2 y neumococo para evitar errores diagnósticos y retrasos en el tratamiento antibiótico


Subject(s)
Humans , Male , Female , Adult , Middle Aged , Aged , Aged, 80 and over , Superinfection/complications , Pneumococcal Infections/pathology , Coronavirus Infections/diagnosis , Betacoronavirus , Pneumonia, Viral/diagnosis , Pneumococcal Infections/drug therapy , Coronavirus Infections/complications , Coronavirus Infections/drug therapy , Pneumonia, Viral/complications , Pneumonia, Viral/drug therapy , Polymerase Chain Reaction , Anti-Bacterial Agents/administration & dosage , Radiography, Thoracic
SELECTION OF CITATIONS
SEARCH DETAIL