10 research outputs found

    First reported genome of an mcr-9-mediated colistin-resistant Salmonella Typhimurium isolate from Brazilian livestock

    Get PDF
    Objectives: To investigate the genetic context of colistin resistance in anmcr-9-harbouring Salmonella Typhimurium ST19 strain from swine in Brazil. Methods: Minimum inhibitory concentrations (MIC) to colistin were determined by broth microdilution. Whole-genome sequencing was performed on an Illumina MiSeq system, followed by de novo genome assembly using SPAdes 1.13.1. The draft genome sequence was annotated in Prokka using KBase online server. Downstream analyses for resistome and plasmid detection were performed using online tools available at the Center for Genomic Epidemiology. The strain was typed in silico using MLST 2.0. Phylogenetic analysis involving 24 other genomes of Salmonella Typhimurium ST19 and mcr-9-harbouring Salmonella Typhimurium isolated from humans, livestock and foodstuff in different regions was also performed. Results: Assembly of the draft genome resulted in 5245 protein-coding sequences, 14 rRNAs, 83 tRNAs and a GC content of 51.81%. The strain was identified as Salmonella Typhimurium ST19 harbouring a 265.5-kb pN1566-2 plasmid carrying genes encoding resistance to colistin (mcr-9.1), aminoglycosides (aadA1), tetracycline [tet(C)] and sulfonamides (sul1). Our findings indicate that the Salmonella Typhimurium ST19 strain in this study showed low genetic variability compared with Salmonella Typhimurium ST19 isolated from swine and poultry in Brazil, and was less related to those reported in other countries. Conclusions: This is the first reported genome of a phenotypically colistin-resistant Salmonella Typhimurium harbouring the mcr-9 variant in Brazilian livestock. This genome will aid global investigations on epidemiological and evolutionary aspects of plasmid-mediated colistin resistance and the role of colistin-resistant Salmonella Typhimurium ST19 lineage as a zoonotic pathogen

    Involvement of the inflammasome in the response of host cells during Staphylococcus aureus infection

    No full text
    Submitted by Izabela Soares ([email protected]) on 2019-10-04T16:50:20Z No. of bitstreams: 2 license_rdf: 811 bytes, checksum: cfd6801dba008cb6adbd9838b81582ab (MD5) Tese de Doutorado_Elma_Lima_Leite.pdf: 9027121 bytes, checksum: 2358a46ca67ad417a77529749f25e3b9 (MD5)Approved for entry into archive by Izabel Miranda ([email protected]) on 2019-10-14T17:41:38Z (GMT) No. of bitstreams: 2 license_rdf: 811 bytes, checksum: cfd6801dba008cb6adbd9838b81582ab (MD5) Tese de Doutorado_Elma_Lima_Leite.pdf: 9027121 bytes, checksum: 2358a46ca67ad417a77529749f25e3b9 (MD5)Made available in DSpace on 2019-10-17T14:11:49Z (GMT). No. of bitstreams: 2 license_rdf: 811 bytes, checksum: cfd6801dba008cb6adbd9838b81582ab (MD5) Tese de Doutorado_Elma_Lima_Leite.pdf: 9027121 bytes, checksum: 2358a46ca67ad417a77529749f25e3b9 (MD5) Previous issue date: 2019-08-20CAPES - Coordenação de Aperfeiçoamento de Pessoal de Nível SuperiorOutra AgênciaStaphylococcus aureus (S. aureus) é uma bactéria Gram-positiva altamente adaptativa e versátil que pode causar uma ampla gama de doenças infecciosas em humanos ou animais. Nos estágios iniciais da infecção, a interação entre S. aureus e as células hospedeiras causa inflamação, cujo processo depende de vários mecanismos celulares complexos e de uma cascata de sinalização coordenada principalmente pelas citocinas e sua ativação. Os principais fatores que determinam o início da inflamação e sua progressão são pathogen- /microbe-associated molecular patterns (PAMPs/ MAMPs), danger-associated molecular patterns (DAMPs), pattern recognition receptors (PRRs), e o sistema imune inato e adaptativo. Inflammassomas, complexos de sinalização multiprotéicos que se agrupam após a detecção de PAMPs/DAMPs, são fatores-chave da resposta imune inata. A maioria dos inflamassomas é formado por um receptor NLR, uma proteína adaptadora ASC e uma Caspase-1 ativa. Uma vez montados, os inflamassomas atuam como plataformas ativadoras que promovem a maturação das citocinas pró-inflamatórias IL-1β e IL-18 em suas formas ativas. Seu papel em diferentes tipos de fagócitos profissionais durante a infecção por S. aureus tem sido extensivamente estudado. Por outro lado, o conhecimento sobre o envolvimento de inflamassomas em fagócitos não profissionais (por exemplo, células epiteliais, células endoteliais ...) é muito fragmentado. O objetivo deste trabalho foi estudar o papel da ativação do inflamassoma durante a infecção por S. aureus em fagócitos não profissionais. Adotamos um modelo de infecção por S. aureus desenvolvido em células osteoblásticas (linhagem MG- 63). Análises de Western blot e ELISA foram usadas para controlar a ativação de caspase-1 e a maturação de IL-1β na interação de S. aureus/MG-63. Geramos células MG-63 deletadas do gene da caspase-1 (CASP1–/– MG-63) usando a abordagem CRISPR/Cas9. Isso nos permitiu demonstrar o envolvimento do inflamassoma na resposta imune inata a S. aureus por fagócitos não profissionais. Inesperadamente, mostramos que S. aureus prolifera nas células CASP1–/– MG-63, sugerindo que Casp1 (ou um fenômeno regulado pela Casp1) está envolvido na eliminação do S. aureus intracelular. Descobrimos que a formação do inflamassoma nas células MG-63 é mais tardia que nos fagócitos profissionais. Além disso, o uso de cepas de S. aureus (mutantes deletados) expressando ou não Phenol-Soluble Modulins (PSM) nos permitiu determinar o papel das PSMs como desencadeador. Assim, demonstramos que S. aureus é capaz de ativar a formação de inflamassomas em células não fagocíticas (MG-63) e que os PSMs estão envolvidos nesse fenômeno.Staphylococcus aureus (S. aureus) is a highly adaptive and versatile Gram-positive bacterium that can cause a wide range of infectious diseases in humans or animals. In the early stages of infection, the interaction between S. aureus and the host cells causes inflammation, the process of which depends on several complex cellular mechanisms and a signaling cascade coordinated mainly by cytokines and their activation. The key factors determining the initiation of inflammation and its progression are pathogen/ microbe-associated molecular patterns (PAMPs/MAMPs), danger-associated molecular patterns (DAMPs), pattern recognition receptors (PRRs), innate and adaptive immunity. Inflammasomes, multi-protein signaling complexes that assemble after sensing PAMPs/ DAMPs, are key players of the innate immune response. Most inflammasomes are formed by an NLR receptor, an ASC adapter protein, and active caspase-1. Once assembled, inflammasomes act as activating platforms that promote the maturation of pro-inflammatory cytokines IL-1β and IL-18 in their active forms. Their role in different types of professional phagocytes during S. aureus infection has been extensively studied. Conversely, knowledge about the involvement of inflammasomes in non-professional phagocytes (e.g. epithelial cells, endothelial cells...) is very fragmented. This work aimed to study the role of inflammasome activation during S. aureus infection in non-occupational phagocytes. We adopted a model of S. aureus infection developed on osteoblastic cells (MG- 63 line). Western blot and ELISA analyses were used to control caspase-1 activation and IL- 1β maturation in the S. aureus MG-63 interaction. We generated MG-63 cells deleted from the caspase-1 gene (CASP1–/– MG-63 cells) using the CRISPR/Cas9 approach. This allowed us to demonstrate the involvement of inflammasomes in the innate immune response to S. aureus by non-professional phagocytes. Unexpectedly, we have shown that S. aureus proliferates in CASP1–/– MG-63 cells, suggesting that Casp1 (or Casp1-regulated phenomenon) is involved in the elimination of intracellular S. aureus. We found that the formation of the inflammasome in MG-63 cells is later than in professional phagocytes. Besides, the use of S. aureus strains (deletion mutants) expressing or not Phenol-Soluble Modulins (PSM) allowed us to determine the role of PSM in this trigger. Thus, we have demonstrated that S. aureus can activate inflammasome formation in non-professional phagocytes (MG-63) cells and that PSM is involved in this phenomenon

    Involvement of the inflammasome in the response of host cells during <em>Staphylococcus aureus</em> infection

    No full text
    Staphylococcus aureus (S. aureus) is a highly adaptive and versatile Gram-positive bacterium that can cause a wide range of infectious diseases in humans or animals. In the early stages of infection, the interaction between S. aureus and the host cells causes inflammation, the process of which depends on several complex cellular mechanisms and a signaling cascade coordinated mainly by cytokines and their activation. The key factors determining the initiation of inflammation and its progression are pathogen/ microbe-associated molecular patterns (PAMPs/MAMPs), danger-associated molecular patterns (DAMPs), pattern recognition receptors (PRRs), innate and adaptive immunity. Inflammasomes, multi-protein signaling complexes that assemble after sensing PAMPs/ DAMPs, are key players of the innate immune response. Most inflammasomes are formed by an NLR receptor, an ASC adapter protein, and active caspase-1. Once assembled, inflammasomes act as activating platforms that promote the maturation of pro-inflammatory cytokines IL-1β and IL-18 in their active forms. Their role in different types of professional phagocytes during S. aureus infection has been extensively studied. Conversely, knowledge about the involvement of inflammasomes in non-professional phagocytes (e.g. epithelial cells, endothelial cells...) is very fragmented. This work aimed to study the role of inflammasome activation during S. aureus infection in non-occupational phagocytes. We adopted a model of S. aureus infection developed on teoblastic cells (MG-63 line). Western blot and ELISA analyses were used to control caspase-1 activation and IL-1β maturation in the S. aureus MG-63 interaction. We generated MG-63 cells deleted from the caspase-1 gene (CASP1–/– MG-63 cells) using the CRISPR/Cas9 approach. This allowed us to demonstrate the involvement of inflammasomes in the innate immune response to S. aureus by non-professional phagocytes. Unexpectedly, we have shown that S. aureus proliferates in CASP1–/– MG-63 cells, suggesting that Casp1 (or Casp1-regulated phenomenon) is involved in the elimination of intracellular S. aureus. We found that the formation of the inflammasome in MG-63 cells is later than in professional phagocytes. Besides, the use of S. aureus strains (deletion mutants) expressing or not Phenol-Soluble Modulins (PSM) allowed us to determine the role of PSM in this trigger. Thus, we have demonstrated that S. aureus can activate inflammasome formation in non-professional phagocytes (MG-63) cells and that PSM is involved in this phenomenon.Staphylococcus aureus (S. aureus) est une bactérie à Gram positif hautement adaptative et polyvalente qui peut causer un large éventail de maladies infectieuses chez l’homme ou l’animal. Lors des premières étapes de l’infection, l’interaction entre S. aureus et les cellules hôtes entraine une inflammation dont le processus dépend de plusieurs mécanismes cellulaires complexes et une cascade de signalisation coordonnée principalement par des cytokines et leur activation. Les facteurs clés déterminant l’initiation de l’inflammation et sa progression sont les pathogen-/microbe-associated molecular patterns (PAMPs/MAMPs), les danger-associated molecular patterns (DAMPs), les Pattern Recognition Receptors (PRRs), l’immunité innée et adaptative. Les inflammasomes, complexes de signalisation multiprotéines qui s'assemblent après la détection de PAMPs/DAMPs sont des facteurs clés de la réponse immunitaire innée. La plupart des inflammasomes sont composés d’un récepteur NLR, d’une protéine adaptatrice ASC et de la caspase-1 activée. Une fois assemblés, les inflammasomes agissent comme des plateformes activatrices qui favorisent la maturation des cytokines proinflammatoires IL-1β et IL-18 sous leurs formes actives. Leur rôle dans les différents types de phagocytes professionnels au cours de l'infection à S. aureus a été largement étudié. A l’inverse, les connaissances sur l'implication des inflammasomes dans les phagocytes nonprofessionnels (e.g. cellules épithéliales, endothéliales…) sont très parcellaires. L'objectif de ce travail était d'étudier le rôle de l'activation de l'inflammasome lors d’une infection à S. aureus sur des phagocytes non-professionnels. Nous avons adopté pour cela un modèle d'infection à S. aureus mis au point sur des cellules ostéoblastiques (lignée MG-63). Des analyses Western blot et ELISA ont été utilisées pour contrôler l'activation de Caspase-1 et la maturation de l’IL-1β lors de l’interaction S. aureus MG-63. Nous avons généré des cellules MG-63 délétées du gène caspase-1 (CASP1–/– MG-63 cells) en utilisant l'approche CRISPR/Cas9. Ceci nous a permis de démontrer l’implication des inflammasomes dans la réponse immune innée à S.aureus par des phagocytes non-professionnels. De façon inattendue, nous avons montré que S. aureus prolifère dans les CASP1–/– MG-63 cells, suggérant que Casp1 (ou les phénomènes régulés par Casp1) est impliquée dans l’élimination de S. aureus intracellulaire. Nous avons constaté que la formation de l'inflammasome dans les cellules MG-63 est cependant plus tardive que dans des phagocytes professionnels. De plus, l'utilisation de souches de S. aureus (mutants de délétion) exprimant ou non des Phenol Soluble Modulins (PSM) nous a permis de déterminer le rôle des PSM dans ce déclenchement. Ainsi, nous avons démontré que S.aureus est capable d’activer la formation d’inflammasome dans des cellules (MG-63), phagocytes non professionnels et que les PSM sont impliqués dans ce phénomène

    Inflammasomes involvement in Staphylococcus aureus infection of human osteoblast-like cells MG-63

    No full text
    Inflammation is a coordinated immune response to infections or tissue damage. The inflammasome is a multi-protein signaling platform that assembles after recognition of danger signals and/or pathogens by a family of cytosolic receptors called NLRs (nucleotide-binding domain and leucine rich repeats containing receptors) or PYHIN protein family members. Once assembled, inflammasomes initiate signaling by activation of downstream proteases, most notably Caspase-1 and Caspase-11, which then proteolytically mature pro-IL-1β, pro-IL-18, and pro-IL-33, and promote their secretion from the cell. Furthermore, inflammasome activation triggers pyroptosis, an inflammatory form of cell death. Staphylococcus aureus is a highly adaptive and versatile gram-positive bacterium that has major importance to human and animal health. S. aureus can cause life-threatening infections such as bacteremia, pneumonia, meningitis, endocarditis and sepsis. S. aureus are also the predominant cause of bone infections worldwide. Comprehending the mechanisms by which staphylococci interact with and damage bone is critical to the development of new approaches to meet this challenge. While the role of inflammasomes formed in the different types of phagocytes during S. aureus infection was widely investigated, the involvement of inflammasomes in osteoblast cells have not been studied. Objective: To understand the mechanisms of Bone Joint Infection we investigated the involvement of inflammasomes in the model of persistent infection of human osteoblast-like cells.Materials and Methods: CRISPR/Cas9 technology was used to prepare Caspase-1 deficient line of MG-63 cells. Western blot analysis and ELISA were employed for the detection of activated cytokines.Results: An employment of wild type vs Caspase-1 deficient MG-63 osteoblast-like cells allow demonstrating the involvement of inflammasomes during S. aureus infection. Using deletion mutants and complemented S. aureus strains, we determined the role of its most important virulence factors for their capacity to activate Caspase-1 and produce IL-1β.Conclusions: Non-phagocytic osteoblast-like MG-63 cells form inflammasomes in the response to S. aureus infection, however the time of inflammasomes formation was different compared to the professional phagocytes. Many virulence factors induce the inflammasomes assemblage

    Inflammasome activation and an involvement of Caspase-1 in a bacterial clearance during S. aureus infection.

    No full text
    International audienceStaphylococcus aureus is a highly versatile Gram-positive bacterium that is carried asymptomatically by up to fifty percent of healthy people, while being a major cause of hospital-acquired infections including bone and joint infections, such as osteomyelitis, which are prone to recurrence. S. aureus-caused bovine mastitis is also significant problem in veterinary medicine. The innate immune response plays a pivotal role in the defense against S. aureus. This response is initiated through pattern recognition receptors, and involves an activation of multi-protein signaling complexes known as inflammasomes. It activates proteases, most notably caspase-1 that proteolytically matures and promotes the secretion of mature IL-1β and IL-18. We investigated the role of inflammasomes and caspase-1 in the secretion of mature IL-1β and in the defence of S. aureus-infected osteoblast-like MG-63 cells. Using CRISPR-Cas9 technology, we demonstrated that MG-63 but not caspase-1 knock-out CASP1−/−MG-63 cells, activate the inflammasome as monitored by the release of mature IL-1β. The use of S. aureus deletion and complemented phenole soluble modulins (PSMs) mutants demonstrated a key role of PSMs in inflammasomes-related IL-1β production. Furthermore, we investigated the role of caspase-1in S. aureus clearance. We found that the lack of caspase-1 in CASP1−/−MG-63 cells impairs their defense functions, as bacterial clearance was drastically decreased in CASP1−/− MG-63 compared to wild-type cells.Using a flow cytometric approach we isolated only S. aureus-bearing cells from mixed populations that allows to identify signals specific to intracellular infection. After RNA-seq and KEGG and Reactome pathway enrichment analysis, the remodeled transcriptomic profile of infected cells revealed exacerbated immune and inflammatory responses, as well as metabolic and epigenetic dysregulations that likely influence the intracellular life of bacteria. Collectively, our results demonstrate that human osteoblast-like cells induce an immune response against S. aureus through inflammasomes activation and processing of IL-1β. The outcome of the infection depends on the balance between the host immune response and the action of main S. aureus virulence factors, such as PSMs, whose production differ among the S. aureus strains. Our results showed that the active caspase-1 prevents exacerbated intracellular replication of S. aureus in non-professional phagocytes in addition to professional phagocytes, suggesting the crucial role of caspase-1 in S. aureus clearance independently from the type of cells. Furthermore, our results provide an atlas of deregulated host genes and biological pathways and identify novel markers and potential candidates for prophylactic and therapeutic approaches

    Involvement of caspase-1 in inflammasomes activation and bacterial clearance in S. aureus-infected osteoblast-like MG-63 cells

    No full text
    International audienceStaphylococcus aureus, a versatile Gram-positive bacterium, is the main cause of bone and joint infections (BJI), which are prone to recurrence. The inflammasome is an immune signaling platform that assembles after pathogen recognition. It activates proteases, most notably caspase-1 that proteolytically matures and promotes the secretion of mature IL-1 beta and IL-18. The role of inflammasomes and caspase-1 in the secretion of mature IL-1 beta and in the defence of S. aureus-infected osteoblasts has not yet been fully investigated. We show here that S. aureus-infected osteoblast-like MG-63 but not caspase-1 knock-out CASP1( -/-)MG-63 cells, which were generated using CRISPR-Cas9 technology, activate the inflammasome as monitored by the release of mature IL-1 beta. The effect was strain-dependent. The use of S. aureus deletion and complemented phenole soluble modulins (PSMs) mutants demonstrated a key role of PSMs in inflammasomes-related IL-1 beta production. Furthermore, we found that the lack of caspase-1 in CASP1( -/-)MG-63 cells impairs their defense functions, as bacterial clearance was drastically decreased in CASP1( -/-) MG-63 compared to wild-type cells. Our results demonstrate that osteoblast-like MG-63 cells play an important role in the immune response against S. aureus infection through inflammasomes activation and establish a crucial role of caspase-1 in bacterial clearance

    Inflammasome activation and an involvement of Caspase-1 in a bacterial clearance during S. aureus infection.

    No full text
    International audienceStaphylococcus aureus is a highly versatile Gram-positive bacterium that is carried asymptomatically by up to fifty percent of healthy people, while being a major cause of hospital-acquired infections including bone and joint infections, such as osteomyelitis, which are prone to recurrence. S. aureus-caused bovine mastitis is also significant problem in veterinary medicine. The innate immune response plays a pivotal role in the defense against S. aureus. This response is initiated through pattern recognition receptors, and involves an activation of multi-protein signaling complexes known as inflammasomes. It activates proteases, most notably caspase-1 that proteolytically matures and promotes the secretion of mature IL-1β and IL-18. We investigated the role of inflammasomes and caspase-1 in the secretion of mature IL-1β and in the defence of S. aureus-infected osteoblast-like MG-63 cells. Using CRISPR-Cas9 technology, we demonstrated that MG-63 but not caspase-1 knock-out CASP1−/−MG-63 cells, activate the inflammasome as monitored by the release of mature IL-1β. The use of S. aureus deletion and complemented phenole soluble modulins (PSMs) mutants demonstrated a key role of PSMs in inflammasomes-related IL-1β production. Furthermore, we investigated the role of caspase-1in S. aureus clearance. We found that the lack of caspase-1 in CASP1−/−MG-63 cells impairs their defense functions, as bacterial clearance was drastically decreased in CASP1−/− MG-63 compared to wild-type cells.Using a flow cytometric approach we isolated only S. aureus-bearing cells from mixed populations that allows to identify signals specific to intracellular infection. After RNA-seq and KEGG and Reactome pathway enrichment analysis, the remodeled transcriptomic profile of infected cells revealed exacerbated immune and inflammatory responses, as well as metabolic and epigenetic dysregulations that likely influence the intracellular life of bacteria. Collectively, our results demonstrate that human osteoblast-like cells induce an immune response against S. aureus through inflammasomes activation and processing of IL-1β. The outcome of the infection depends on the balance between the host immune response and the action of main S. aureus virulence factors, such as PSMs, whose production differ among the S. aureus strains. Our results showed that the active caspase-1 prevents exacerbated intracellular replication of S. aureus in non-professional phagocytes in addition to professional phagocytes, suggesting the crucial role of caspase-1 in S. aureus clearance independently from the type of cells. Furthermore, our results provide an atlas of deregulated host genes and biological pathways and identify novel markers and potential candidates for prophylactic and therapeutic approaches

    Unleashing the power of inflammasomes and trained immunity: promising strategies in the fight against Staphylococcus aureus infection

    No full text
    International audienceStaphylococcus aureus causes life-threatening diseases such as pneumonia and osteomyelitis. Pathogens activate inflammasomes that trigger protease activation, particularly caspase-1, which promotes the secretion of mature IL-1β and IL-18. Trained immunity (TI) enhances the immune response to subsequent unrelated challenges through epigenetic reprogramming of transcriptional pathways and alteration of cell metabolism. Our objectives include a comparison of IL-1β production by monocyte–like ThP1 cells vs osteoblast-like MG-63 cells and comprehending the role of inflammasomes effector, caspase-1, investigating the development of TI in non-immune MG-63 and epithelial lung A549 cells, in the context of S. aureus infection.The role of inflammasomes and Caspase-1 was examined by a comparison of MG-63 cells and caspase-1 knock-out CASP1−/−MG-63 cells that were produced using CRISPR-Cas9 technology. We developed an in vitro TI model using MG-63 and A549 cells and investigated the involvement of ROS using the scavenger N-acetylcysteine (NAC). ThP1 cells produced a higher amount of IL-1β compared to MG-63 cells. Infected MG-63 cells release mature IL-1β, while CASP1−/−MG-63 cells didn’t. PSMs were identified as key contributors to IL-1β production using deletion and complemented phenol-soluble modulins (PSMs) S. aureus mutants. Caspase-1 deficiency impaired cell defense, resulting in decreased bacterial clearance in CASP1−/−MG-63 cells. β-glucan training of MG-63 and A549 cells increased IL-6/IL-8 production upon a stimulation with S. aureus. Interleukin production positively correlated with Histone 3 acetylation (H3K27), indicating epigenetic reprogramming. NAC addition, prior to β-glucan training before S. aureus infection, inhibited IL-6/IL-8 production, thereby supporting the involvement of ROS in the induction of TI. Cell exposure to bacterium with probiotic properties, Lactococcus lactis, before S. aureus infection elevated IL-6/IL-8 production, accompanied by H3K27 acetylation, suggesting its ability to induce TI. Our results demonstrate two host strategies against S. aureus infection with therapeutic potential

    Development of trained immunity and activation of inflammasomes are promising strategies to combat Staphylococcus aureus infection

    No full text
    International audienceIntroduction and objectives Staphylococcus aureus causes life-threatening diseases such as pneumonia and osteomyelitis. Trained immunity (TI) enhances the immune response to subsequent unrelated challenges through epigenetic reprogramming of transcriptional pathways and alteration of cell metabolism [1]. Moreover, pathogens activate inflammasomes that trigger protease activation, particularly caspase-1. Caspase-1 proteolytically matures and promotes the secretion of mature IL-1β and IL-18 [2]. Our objectives include investigating the development of TI in non-immune cells, examining interleukin production and epigenetic marks, exploring the involvement of reactive oxygen species (ROS), and comprehending the role of inflammasomes and their effector, caspase-1, in the context of S. aureus infection.Materials and methodsWe developed an in vitro TI model using human osteoblast-like MG-63 and epithelial lung A549 cells [3]. Techniques employed included microbiology methods of bacterial cultivation, enzyme-linked immunosorbent assay (ELISA), microscopic analysis, and flow cytometry. We investigated the involvement of ROS using the scavenger N-acetylcysteine (NAC) [4]. The role of inflammasomes and Caspase-1 was examined by a comparison of MG-63 cells and caspase-1 knock-out CASP1−/−MG-63 cells that were generated via CRISPR-Cas9 technology [5]. The role of bacterial effectors were analyzed using deletion and complemented phenol-soluble modulins (PSMs) mutants.Results, discussion and conclusion β-glucan training of MG-63 and A549 cells increased IL-6/IL-8 production upon a stimulation with S. aureus. Interleukin production positively correlated with Histone 3 acetylation at lysine 27 (H3K27), indicating epigenetic reprogramming. NAC addition, prior to β-glucan training before S. aureus infection, inhibited IL-6/IL-8 production, thereby supporting the involvement of ROS in the induction of TI. Cell exposure to bacterium with probiotic properties, Lactococcus lactis, before S. aureus infection also elevated IL-6/IL-8 production, accompanied by H3K27 acetylation, suggesting its ability to induce TI [3]. Infected MG-63 cells activated inflammasomes, releasing mature IL-1β, while CASP1−/−MG-63 cells lacked this activation. PSMs were identified as key contributors to inflammasome-related IL-1β production using S. aureus mutants. Additionally, caspase-1 deficiency impaired cell defense, resulting in decreased bacterial clearance in CASP1−/−MG-63 cells [5]. Our results demonstrate two host strategies against S. aureus infection with therapeutic potential and reveal the involvement of non-immune cells in defense response beyond their structural functions and tissue homeostasis maintenance

    Lifelong robbery victimisation and mental disorders at age 18 years: Brazilian population-based study

    No full text
    Purpose: Urban violence is a major problem in Brazil and may contribute to mental disorders among victims. The aim of this study was to assess the association between robbery victimisation and mental health disorders in late adolescence. Methods: At age 18 years, 4106 participants in the 1993 Pelotas Birth Cohort Study were assessed. A questionnaire about history of robbery victimisation was administered, the Self-Report Questionnaire was used to screen for common mental disorders, and the Mini International Neuropsychiatric Interview was used to assess major depressive disorder and generalised anxiety disorder. Cross-sectional prevalence ratios between lifetime robbery victimisation and mental disorders were estimated using Poisson regression with robust standard errors, adjusting for socioeconomic variables measured at birth and violence in the home and maltreatment measured at age 15. Results: There was a dose–response relationship between frequency of lifetime robberies and risk of mental disorders. Adolescents who had been robbed three or more times had twice the risk (PR 2.04; 95% CI 1.64–2.56) for common mental disorders, over four times the risk for depression (PR 4.59; 95% CI 2.60–8.12), and twice the risk for anxiety (PR 1.93; 95% CI 1.06–3.50), compared with non-victims, adjusting for covariates. Experiencing frequent robberies had greater impact on common mental disorders than experiencing an armed robbery. Population attributable fractions with regard to robbery were 9% for common mental disorders, 13% for depression, and 8% for anxiety. Conclusions: Robberies are associated with common mental disorders in late adolescence, independently of violence between family members. Reducing urban violence could significantly help in preventing common mental illnesses
    corecore