Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-38370826

RESUMO

Toll-like receptor 9 (TLR9) is an innate immune receptor that localizes to endosomes in antigen presenting cells and recognizes single stranded unmethylated CpG sites on bacterial genomic DNA. Previous bioinformatic studies have indicated that the genome of the human pathogen Chlamydia trachomatis contains TLR9 stimulatory motifs, and correlative studies have implied a link between human TLR9 (hTLR9) genotype variants and susceptibility to infection. Here we present our evaluation of the stimulatory potential of C. trachomatis gDNA and its recognition by hTLR9- and murine TLR9 (mTLR9)-expressing cells. We confirm that hTLR9 colocalizes with chlamydial inclusions in the pro-monocytic cell line, U937. Utilizing HEK293 reporter cell lines, we demonstrate that purified genomic DNA from C. trachomatis can stimulate hTLR9 signaling, albeit at lower levels than gDNA prepared from other Gram-negative bacteria. Interestingly, we found that while C. trachomatis is capable of signaling through hTLR9 and mTLR9 during live infections in non-phagocytic HEK293 reporter cell lines, signaling only occurs at later developmental time points. Chlamydia-specific induction of hTLR9 is blocked when protein synthesis is inhibited prior to the RB-to-EB conversion and exacerbated by the inhibition of lipooligosaccharide biosynthesis. The induction of aberrance / persistence also significantly alters Chlamydia-specific TLR9 signaling. Our observations support the hypothesis that chlamydial gDNA is released at appreciable levels by the bacterium during the conversion between its replicative and infectious forms and during treatment with antibiotics targeting peptidoglycan assembly.

2.
Infect Immun ; 91(11): e0023623, 2023 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-37861311

RESUMO

Clostridioides difficile is the most common cause of nosocomial gastrointestinal tract bacterial infections. We lack fully effective reliable treatments for this pathogen, and there is a critical need to better understand how C. difficile interacts with our immune system. Group 3 innate lymphocytes (ILC3s) are rare immune cells localized within mucosal tissues that protect against bacterial infections. Upon activation, ILC3s secrete high levels of the cytokine interleukin-22 (IL-22), which is a critical regulator of tissue responses during infection. C. difficile toxin B (TcdB), the major virulence factor, directly activates ILC3s, resulting in high IL-22 levels. We previously reported that polyamines are important in the activation of ILC3s by the innate cytokine interleukin-23 (IL-23) but did not identify a specific mechanism. In this study, we examine how a pathogen impacts a metabolic pathway important for immune cell function and hypothesized that polyamines are important in TcdB-mediated ILC3 activation. We show that TcdB upregulates the polyamine biosynthesis pathway, and the inhibition of the pathway decreases TcdB-mediated ILC3 activation. Two polyamines, putrescine and spermidine, are involved. Spermidine is the key polyamine in the hypusination of eukaryotic initiation factor 5A (eIF5A), and the inhibition of eIF5A reduced ILC3 activation. Thus, there is potential to leverage polyamines in ILC3s to promote activation of ILC3s during C. difficile infection and other bacterial infections where ILC3s serve a protective role.


Assuntos
Infecções Bacterianas , Toxinas Bacterianas , Clostridioides difficile , Gastroenteropatias , Humanos , Toxinas Bacterianas/metabolismo , Poliaminas/metabolismo , Espermidina/metabolismo , Linfócitos , Proteínas de Bactérias/metabolismo , Citocinas/metabolismo
3.
Front Immunol ; 14: 1219072, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37483638

RESUMO

Innate lymphoid cells (ILCs) are key regulators of tissue homeostasis, inflammation, and immunity to infections. ILCs rapidly respond to environmental cues such as cytokines, microbiota and invading pathogens which regulate their function and phenotype. Even though ILCs are rare cells, they are enriched at barrier surfaces such as the gastrointestinal (GI) tract, and they are often critical to the host's immune response to eliminate pathogens. On the other side of host-pathogen interactions, pathogenic bacteria also have the means to modulate these immune responses. Manipulation or evasion of the immune cells is often to the pathogen's benefit and/or to the detriment of competing microbiota. In some instances, specific bacterial virulence factors or toxins have been implicated in how the pathogen modulates immunity. In this review, we discuss the recent progress made towards understanding the role of non-cytotoxic ILCs during enteric bacterial infections, how these pathogens can modulate the immune response, and the implications these have on developing new therapies to combat infection.


Assuntos
Microbioma Gastrointestinal , Imunidade Inata , Humanos , Linfócitos , Inflamação , Trato Gastrointestinal , Enterobacteriaceae , Bactérias
4.
Immunohorizons ; 7(1): 41-48, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36637514

RESUMO

Group 3 innate lymphocytes (ILC3s) rapidly respond to invading pathogens or inflammatory signals, which requires shifting cellular metabolic demands. Metabolic adaptations regulating ILC3 function are not completely understood. Polyamines are polycationic metabolites that have diverse roles in cellular functions and in immunity regulate immune cell biology, including Th17 cells. Whether polyamines play a role in ILC3 activation is unknown. In this article, we report that the polyamine synthesis pathway is important for ILC3 activation. IL-23-activated mouse ILC3s upregulate ornithine decarboxylase, the enzyme catalyzing the rate-limiting step of the conversion of ornithine to putrescine in polyamine synthesis, with a subsequent increase in putrescine levels. Inhibition of ornithine decarboxylase via a specific inhibitor, α-difluoromethylornithine, reduced levels of IL-22 produced by steady-state or IL-23-activated ILC3s in a putrescine-dependent manner. Thus, the polyamine putrescine is a positive regulator of ILC3 activation. Our results suggest that polyamines represent a potential target for therapeutic modulation of ILC3 activation during infection or inflammatory disorders.


Assuntos
Poliaminas , Putrescina , Camundongos , Animais , Poliaminas/metabolismo , Poliaminas/farmacologia , Putrescina/farmacologia , Ornitina Descarboxilase/metabolismo , Ativação Linfocitária , Interleucina-23
5.
Infect Immun ; 90(4): e0007322, 2022 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-35377172

RESUMO

Group 3 innate lymphocytes (ILC3s) are rare immune cells localized in mucosal tissues, especially the gastrointestinal (GI) tract. Despite their rarity, they are a major source of the cytokine interleukin-22 (IL-22), which protects the GI epithelium during inflammation and infection. Although ILC3s have been demonstrated to be important for defense against Clostridioides difficile infection, the exact mechanisms through which they sense productive infection and become activated to produce IL-22 remain poorly understood. In this study, we identified a novel mechanism of ILC3 activation after exposure to C. difficile. Toxin B (TcdB) from C. difficile directly induced production of IL-22 in ILC3s, and this induction was dependent on the glucosyltransferase activity of the toxin, which inhibits small GTPases. Pharmacological inhibition of the small GTPase Cdc42 also enhanced IL-22 production in ILC3s, indicating that Cdc42 is a negative regulator of ILC3 activation. Further gene expression analysis revealed that treatment with TcdB modulated the expression of several inflammation-related genes in ILC3s. These findings demonstrate that C. difficile toxin-mediated inhibition of Cdc42 leads to the activation of ILC3s, providing evidence for how these cells are recruited into the immune response against the pathobiont.


Assuntos
Toxinas Bacterianas , Clostridioides difficile , Infecções por Clostridium , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Humanos , Imunidade Inata , Inflamação/metabolismo , Linfócitos
6.
Cytokine ; 153: 155862, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35306427

RESUMO

Group 3 innate lymphocytes (ILC3s) are important immune cells within mucosal tissues and protect against bacterial infections. They can be activated in response to the innate cytokines IL-23 or IL-1ß, which rapidly increases their production of effector molecules that regulate barrier functions. Pathogens can subvert these anti-bacterial effects to evade mucosal defenses to infect the host. Bacillus anthracis, the causative agent of anthrax, produces two major toxins that can modulate the immune response. We have previously shown that lethal toxin downmodulates the function of ILC3s. On the other hand, edema toxin has been shown promote T helper 17 (Th17) cell differentiation, adaptive counterparts of ILC3s, via elevation of cyclic adenosine monophosphate (cAMP). We hypothesized that edema toxin may also modulate ILC3 function. In this study, we show that edema toxin has the opposite effect of lethal toxin; edema toxin directly activates ILC3s independently of innate cytokine stimulation. Treatment of a mouse ILC3-like cell line with edema toxin, a potent adenylate cyclase, upregulated production of the cytokine IL-22, a major effector molecule of ILC3s and a critical factor in maintaining mucosal barriers. Forskolin treatment phenocopied the effect observed with edema toxin and led to an increase in CREB phosphorylation in ILC3s. This observation has potential implications for a role for cAMP signaling in the activation of ILC3s.


Assuntos
Toxinas Bacterianas , Animais , Antígenos de Bactérias , Toxinas Bacterianas/farmacologia , AMP Cíclico , Citocinas , Edema , Imunidade Inata , Interleucinas , Linfócitos , Camundongos
7.
ACS Appl Bio Mater ; 5(2): 545-551, 2022 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-35113537

RESUMO

The reliance on antibiotics and antimicrobials to treat bacterial infectious diseases is threatened by the emergence of antibiotic resistance and multi-drug-resistant organisms, thus having the potential to greatly impact human health. Thus, the discovery and development of antimicrobials capable of acting on antibiotic-resistant bacteria is a major area of significance in scientific research. Herein, we present the development of a eumelanin-inspired antimicrobial capable of killing methicillin-resistant Staphylococcus aureus (MRSA). By ligating quaternary ammonium-functionalized "arms" to a eumelanin-inspired indole with intrinsic antimicrobial activity, an antimicrobial agent with enhanced activity was prepared. This resulting antimicrobial, EIPE-1, had a minimum inhibitory concentration of 16 µg/mL (17.1 µM) against a clinical isolate of MRSA obtained from an adult cystic fibrosis patient. The biocidal activity occurred within 30 min of exposure and resulted in changes to the bacterial cell surface as visualized with a scanning electron microscope. Taken together, these studies demonstrate that EIPE-1 is effective at killing MRSA.


Assuntos
Anti-Infecciosos , Staphylococcus aureus Resistente à Meticilina , Adulto , Antibacterianos/farmacologia , Anti-Infecciosos/farmacologia , Resistência Microbiana a Medicamentos , Humanos , Testes de Sensibilidade Microbiana
9.
Pathogens ; 9(12)2020 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-33321710

RESUMO

Chlamydia species are causative agents of sexually transmitted infections, blinding trachoma, and animal infections with zoonotic potential. Being an obligate intracellular pathogen, Chlamydia relies on the host cell for its survival and development, subverting various host cell processes throughout the infection cycle. A key subset of host proteins utilized by Chlamydia include an assortment of host kinase signaling networks which are vital for many chlamydial processes including entry, nutrient acquisition, and suppression of host cell apoptosis. In this review, we summarize the recent advancements in our understanding of host kinase subversion by Chlamydia.

10.
Pathog Dis ; 77(6)2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31647538

RESUMO

Chlamydia trachomatis is a significant pathogen with global and economic impact. As an obligate intracellular pathogen, C. trachomatis resides inside the inclusion, a parasitophorous vacuole, and depends on the host cell for survival and transition through a biphasic development cycle. During infection, C. trachomatis is known to manipulate multiple signaling pathways and recruit an assortment of host proteins to the inclusion membrane, including host kinases. Here, we show recruitment of multiple isoforms of protein kinase C (PKC) including active phosphorylated PKC isoforms to the chlamydial inclusion colocalizing with active Src family kinases. Pharmacological inhibition of PKC led to a modest reduction of infectious progeny production. PKC phosphorylated substrates were seen recruited to the entire periphery of the inclusion membrane. Infected whole cell lysates showed altered PKC phosphorylation of substrates during the course of infection. Assessment of different chlamydial species showed recruitment of PKC and PKC phosphorylated substrates were limited to C. trachomatis. Taken together, PKC and PKC substrate recruitment may provide significant insights into how C. trachomatis manipulates multiple host signaling cascades during infection.


Assuntos
Infecções por Chlamydia/fisiopatologia , Chlamydia trachomatis/metabolismo , Interações Hospedeiro-Patógeno , Proteína Quinase C/metabolismo , Vacúolos/metabolismo , Vacúolos/microbiologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Células HeLa , Humanos , Fosforilação , Processamento de Proteína Pós-Traducional , Transdução de Sinais
11.
Artigo em Inglês | MEDLINE | ID: mdl-30555802

RESUMO

Chlamydia trachomatis is an obligate intracellular pathogen with global health and economic impact. Upon infection, C. trachomatis resides within a protective niche, the inclusion, wherein it replicates and usurps host cell machinery and resources. The inclusion membrane is the key host-pathogen interface that governs specific protein-protein interactions to manipulate host signaling pathways. At the conclusion of the infection cycle, C. trachomatis exits the host cell via lysis or extrusion. Extrusion depends on the phosphorylation state of myosin light chain 2 (MLC2); the extent of phosphorylation is determined by the ongoing opposing activities of myosin phosphatase (MYPT1) and myosin kinase (MLCK). Previously, it was shown that MYPT1 is recruited to the inclusion and interacts with CT228 for regulation of host cell egress. In this study, we generated a targeted chromosomal mutation of CT228 (L2-ΔCT228) using the TargeTron system and demonstrate a loss of MYPT1 recruitment and increase in extrusion production in vitro. Mutation of CT228 did not affect chlamydial growth in cell culture or recruitment of MLC2. Moreover, we document a delay in clearance of L2-ΔCT228 during murine intravaginal infection as well as a reduction in systemic humoral response, relative to L2-wild type. Taken together, the data suggest that loss of MYPT1 recruitment (as a result of CT228 disruption) regulates the degree of host cell exit via extrusion and affects the longevity of infection in vivo.


Assuntos
Infecções por Chlamydia/metabolismo , Chlamydia trachomatis/genética , Chlamydia trachomatis/patogenicidade , Inativação Gênica , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Animais , Infecções por Chlamydia/imunologia , Infecções por Chlamydia/patologia , Modelos Animais de Doenças , Feminino , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Corpos de Inclusão/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Mutação , Cadeias Leves de Miosina , Quinase de Cadeia Leve de Miosina/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Fosforilação , Domínios e Motivos de Interação entre Proteínas , Útero/patologia
12.
Artigo em Inglês | MEDLINE | ID: mdl-26457182

RESUMO

BACKGROUND: Staphylococcus aureus is a frequent cause of infections in both the community and hospital. Methicillin-resistant Staphylococcus aureus continues to be an important nosocomial pathogen and infections are often difficult to manage due to its resistance to multiple antibiotics. Healthcare workers are important source of nosocomial transmission of MRSA. This study aimed to determine the nasal carriage rate of S. aureus and MRSA among healthcare workers at Universal College of Medical Sciences and Teaching Hospital, Nepal and to determine antibiotic susceptibility pattern of the isolates. METHODS: A cross-sectional study involving 204 healthcare workers was conducted. Nasal swabs were collected and cultured on Mannitol salt agar. Mannitol fermenting colonies which were gram positive cocci, catalase positive and coagulase positive were identified as S. aureus. Antibiotic susceptibility test was performed by modified Kirby-Bauer disc diffusion method. Methicillin resistance was detected using cefoxitin disc diffusion method. RESULTS: Of 204 healthcare workers, 32 (15.7 %) were nasal carriers of S. aureus and among them 7 (21.9 %) were carrier of MRSA. Overall nasal carriage rate of MRSA was 3.4 % (7/204). Highest MRSA nasal carriage rate of 7.8 % (4/51) was found among nurses. Healthcare workers of both surgical wards and operating room accounted for 28.6 % (2/7) of MRSA carriers each. Among MRSA isolates inducible clindamycin resistance was observed in 66.7 % (2/3) of erythromycin resistant isolates. CONCLUSIONS: High nasal carriage of S. aureus and MRSA among healthcare workers (especially in surgery ward and operating room) necessitates improved infection control measures to be employed to control MRSA transmission in our setting.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA