Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Immunity ; 45(1): 106-18, 2016 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-27421701

RESUMO

The ATM kinase is a central component of the DNA damage repair machinery and redox balance. ATM dysfunction results in the multisystem disease ataxia-telangiectasia (AT). A major cause of mortality in AT is respiratory bacterial infections. Whether ATM deficiency causes innate immune defects that might contribute to bacterial infections is not known. Here we have shown that loss of ATM impairs inflammasome-dependent anti-bacterial innate immunity. Cells from AT patients or Atm(-/-) mice exhibited diminished interleukin-1ß (IL-1ß) production in response to bacteria. In vivo, Atm(-/-) mice were more susceptible to pulmonary S. pneumoniae infection in a manner consistent with inflammasome defects. Our data indicate that such defects were due to oxidative inhibition of inflammasome complex assembly. This study reveals an unanticipated function of reactive oxygen species (ROS) in negative regulation of inflammasomes and proposes a theory for the notable susceptibility of AT patients to pulmonary bacterial infection.


Assuntos
Ataxia Telangiectasia/genética , Pulmão/imunologia , Infecções Pneumocócicas/imunologia , Streptococcus pneumoniae/imunologia , Animais , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Células Cultivadas , Dano ao DNA , Reparo do DNA , Humanos , Imunidade Inata , Inflamassomos/fisiologia , Interleucina-1beta , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxirredução , Espécies Reativas de Oxigênio/metabolismo
2.
Bioinformatics ; 37(1): 126-128, 2021 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-33367516

RESUMO

SUMMARY: Since its introduction, RNA-Seq technology has been used extensively in studies of pathogenic bacteria to identify and quantify differences in gene expression across multiple samples from bacteria exposed to different conditions. With some exceptions, tools for studying gene expression, determination of differential gene expression, downstream pathway analysis and normalization of data collected in extreme biological conditions is still lacking. Here, we describe ProkSeq, a user-friendly, fully automated RNA-Seq data analysis pipeline designed for prokaryotes. ProkSeq provides a wide variety of options for analysing differential expression, normalizing expression data and visualizing data and results. AVAILABILITY AND IMPLEMENTATION: ProkSeq is implemented in Python and is published under the MIT source license. The pipeline is available as a Docker container https://hub.docker.com/repository/docker/snandids/prokseq-v2.0, or can be used through Anaconda: https://anaconda.org/snandiDS/prokseq. The code is available on Github: https://github.com/snandiDS/prokseq and a detailed user documentation, including a manual and tutorial can be found at https://prokseqV20.readthedocs.io. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.

3.
Infect Immun ; 89(4)2021 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-33495272

RESUMO

Pathogenic Yersinia spp. depend on the activity of a potent virulence plasmid-encoded ysc/yop type 3 secretion system (T3SS) to colonize hosts and cause disease. It was recently shown that Yersinia pseudotuberculosis upregulates the virulence plasmid copy number (PCN) during infection and that the resulting elevated gene dose of plasmid-encoded T3SS genes is essential for virulence. When and how this novel regulatory mechanism is deployed and regulates the replication of the virulence plasmid during infection is unknown. In the present study, we applied droplet digital PCR (ddPCR) to investigate the dynamics of Y. pseudotuberculosis virulence PCN variations and growth rates in infected mouse organs. We demonstrated that both PCN and growth varied in different tissues and over time throughout the course of infection, indicating that the bacteria adapted to discrete microenvironments during infection. The PCN was highest in Peyer's patches and cecum during the clonal invasive phase of the infection, while the highest growth rates were found in the draining mesenteric lymph nodes. In deeper, systemic organs, the PCN was lower and more modest growth rates were recorded. Our study indicates that increased gene dosage of the plasmid-encoded T3SS genes is most important early in the infection during invasion of the host. The described ddPCR approach will greatly simplify analyses of PCN, growth dynamics, and bacterial loads in infected tissues and will be readily applicable to other infection models.


Assuntos
Carga Bacteriana , Variações do Número de Cópias de DNA , Plasmídeos/genética , Infecções por Yersinia pseudotuberculosis/microbiologia , Yersinia pseudotuberculosis/fisiologia , Animais , Proteínas de Bactérias/genética , Modelos Animais de Doenças , Feminino , Camundongos , Especificidade de Órgãos , Reação em Cadeia da Polimerase em Tempo Real , Virulência , Fatores de Virulência/genética , Infecções por Yersinia pseudotuberculosis/diagnóstico
5.
PLoS Pathog ; 11(1): e1004600, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25590628

RESUMO

We recently found that Yersinia pseudotuberculosis can be used as a model of persistent bacterial infections. We performed in vivo RNA-seq of bacteria in small cecal tissue biopsies at early and persistent stages of infection to determine strategies associated with persistence. Comprehensive analysis of mixed RNA populations from infected tissues revealed that Y. pseudotuberculosis undergoes transcriptional reprogramming with drastic down-regulation of T3SS virulence genes during persistence when the pathogen resides within the cecum. At the persistent stage, the expression pattern in many respects resembles the pattern seen in vitro at 26oC, with for example, up-regulation of flagellar genes and invA. These findings are expected to have impact on future rationales to identify suitable bacterial targets for new antibiotics. Other genes that are up-regulated during persistence are genes involved in anaerobiosis, chemotaxis, and protection against oxidative and acidic stress, which indicates the influence of different environmental cues. We found that the Crp/CsrA/RovA regulatory cascades influence the pattern of bacterial gene expression during persistence. Furthermore, arcA, fnr, frdA, and wrbA play critical roles in persistence. Our findings suggest a model for the life cycle of this enteropathogen with reprogramming from a virulent to an adapted phenotype capable of persisting and spreading by fecal shedding.


Assuntos
Análise de Sequência de RNA/métodos , Virulência/genética , Infecções por Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/patogenicidade , Animais , Ceco/imunologia , Ceco/microbiologia , Ceco/patologia , Feminino , Regulação Bacteriana da Expressão Gênica , Genes Bacterianos , Camundongos , Análise em Microsséries , Microbiota/imunologia , RNA Bacteriano/genética , Yersinia pseudotuberculosis/imunologia , Infecções por Yersinia pseudotuberculosis/imunologia , Infecções por Yersinia pseudotuberculosis/patologia
6.
Infect Immun ; 84(12): 3369-3378, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27620724

RESUMO

Neutrophils are essential components of immunity and are rapidly recruited to infected or injured tissue. Upon their activation, neutrophils release granules to the cell's exterior, through a process called degranulation. These granules contain proteins with antimicrobial properties that help combat infection. The enteropathogenic bacterium Yersinia pseudotuberculosis successfully persists as an extracellular bacterium during infection by virtue of its translocation of virulence effectors (Yersinia outer proteins [Yops]) that act in the cytosol of host immune cells to subvert phagocytosis and proinflammatory responses. Here, we investigated the effect of Y. pseudotuberculosis on neutrophil degranulation upon cell contact. We found that virulent Y. pseudotuberculosis was able to prevent secondary granule release. The blocking effect was general, as the release of primary and tertiary granules was also reduced. Degranulation of secondary granules was also blocked in primed neutrophils, suggesting that this mechanism could be an important element of immune evasion. Further, wild-type bacteria conferred a transient block on neutrophils that prevented their degranulation upon contact with plasmid-cured, avirulent Y. pseudotuberculosis and Escherichia coli Detailed analyses showed that the block was strictly dependent on the cooperative actions of the two antiphagocytic effectors, YopE and YopH, suggesting that the neutrophil target structures constituting signaling molecules needed to initiate both phagocytosis and general degranulation. Thus, via these virulence effectors, Yersinia can impair several mechanisms of the neutrophil's antimicrobial arsenal, which underscores the power of its virulence effector machinery.


Assuntos
Degranulação Celular , Neutrófilos/fisiologia , Yersinia pseudotuberculosis/patogenicidade , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Humanos , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/metabolismo , Virulência , Yersinia pseudotuberculosis/fisiologia
7.
Infect Immun ; 82(3): 1181-91, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24379291

RESUMO

The human-pathogenic species of the Gram-negative genus Yersinia preferentially target and inactivate cells of the innate immune defense, suggesting that this is a critical step by which these bacteria avoid elimination and cause disease. In this study, bacterial interactions with dendritic cells, macrophages, and polymorphonuclear neutrophils (PMNs) in intestinal lymphoid tissues during early Yersinia pseudotuberculosis infection were analyzed. Wild-type bacteria were shown to interact mainly with dendritic cells, but not with PMNs, on day 1 postinfection, while avirulent yopH and yopE mutants interacted with PMNs as well as with dendritic cells. To unravel the role of PMNs during the early phase of infection, we depleted mice of PMNs by using an anti-Ly6G antibody, after which we could see more-efficient initial colonization by the wild-type strain as well as by yopH, yopE, and yopK mutants on day 1 postinfection. Dissemination of yopH, yopE, and yopK mutants from the intestinal compartments to mesenteric lymph nodes was faster in PMN-depleted mice than in undepleted mice, emphasizing the importance of effective targeting of PMNs by these Yersinia outer proteins (Yops). In conclusion, escape from interaction with PMNs due to the action of YopH, YopE, and YopK is a key feature of pathogenic Yersinia species that allows colonization and effective dissemination.


Assuntos
Neutrófilos/imunologia , Infecções por Yersinia pseudotuberculosis/imunologia , Yersinia pseudotuberculosis/imunologia , Animais , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Feminino , Intestinos/imunologia , Intestinos/microbiologia , Tecido Linfoide/imunologia , Tecido Linfoide/microbiologia , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Mutação/genética , Mutação/imunologia , Neutrófilos/microbiologia , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/imunologia , Yersinia pseudotuberculosis/genética , Infecções por Yersinia pseudotuberculosis/microbiologia
8.
Infect Immun ; 82(8): 3471-82, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24891107

RESUMO

Yersiniosis is a human disease caused by the bacterium Yersinia pseudotuberculosis or Yersinia enterocolitica. The infection is usually resolved but can lead to postinfectious sequelae, including reactive arthritis and erythema nodosum. The commonly used Yersinia mouse infection model mimics acute infection in humans to some extent but leads to systemic infection and eventual death. Here, we analyzed sublethal infection doses of Y. pseudotuberculosis in mice in real time using bioluminescent imaging and found that infections using these lower doses result in extended periods of asymptomatic infections in a fraction of mice. In a search for the site for bacterial persistence, we found that the cecum was the primary colonization site and was the site where the organism resided during a 115-day infection period. Persistent infection was accompanied by sustained fecal shedding of cultivable bacteria. Cecal patches were identified as the primary site for cecal colonization during persistence. Y. pseudotuberculosis bacteria were present in inflammatory lesions, in localized foci, or as single cells and also in neutrophil exudates in the cecal lumen. The chronically colonized cecum may serve as a reservoir for dissemination of infection to extraintestinal sites, and a chronic inflammatory state may trigger the onset of postinfectious sequelae. This novel mouse model for bacterial persistence in cecum has potential as an investigative tool to unveil a deeper understanding of bacterial adaptation and host immune defense mechanisms during persistent infection.


Assuntos
Ceco/microbiologia , Infecções por Yersinia pseudotuberculosis/microbiologia , Yersinia pseudotuberculosis/fisiologia , Animais , Derrame de Bactérias , Modelos Animais de Doenças , Fezes/microbiologia , Feminino , Medições Luminescentes , Camundongos , Imagem Corporal Total
9.
Cell Microbiol ; 15(7): 1088-110, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23279117

RESUMO

Type III secretion enables bacteria to intoxicate eukaryotic cells with anti-host effectors. A class of secreted cargo are the two hydrophobic translocators that form a translocon pore in the host cell plasma membrane through which the translocated effectors may gain cellular entry. In pathogenic Yersinia, YopB and YopD shape this translocon pore. Here, four in cis yopD mutations were constructed to disrupt a predicted α-helix motif at the C-terminus. Mutants YopD(I262P) and YopD(K267P) poorly localized Yop effectors into target eukaryotic cells and failed to resist uptake and killing by immune cells. These defects were due to deficiencies in host-membrane insertion of the YopD-YopB translocon. Mutants YopDA(263P) and YopD(A270P) had no measurable in vitro translocation defect, even though they formed smaller translocon pores in erythrocyte membranes. Despite this, all four mutants were attenuated in a mouse infection model. Hence, YopD variants have been generated that can spawn translocons capable of targeting effectors in vitro, yet were bereft of any lethal effect in vivo. Therefore, Yop translocators may possess other in vivo functions that extend beyond being a portal for effector delivery into host cells.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Sistemas de Secreção Bacterianos , Yersinia pseudotuberculosis/metabolismo , Yersinia pseudotuberculosis/patogenicidade , Animais , Proteínas da Membrana Bacteriana Externa/genética , Linhagem Celular , Análise Mutacional de DNA , Modelos Animais de Doenças , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Virulência , Yersiniose/microbiologia , Yersiniose/patologia , Yersinia pseudotuberculosis/genética
10.
Proc Natl Acad Sci U S A ; 108(4): 1639-44, 2011 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-21220342

RESUMO

Pathogenic Yersinia species suppress the host immune response by using a plasmid-encoded type III secretion system (T3SS) to translocate virulence proteins into the cytosol of the target cells. T3SS-dependent protein translocation is believed to occur in one step from the bacterial cytosol to the target-cell cytoplasm through a conduit created by the T3SS upon target cell contact. Here, we report that T3SS substrates on the surface of Yersinia pseudotuberculosis are translocated into target cells. Upon host cell contact, purified YopH coated on Y. pseudotuberculosis was specifically and rapidly translocated across the target-cell membrane, which led to a physiological response in the infected cell. In addition, translocation of externally added YopH required a functional T3SS and a specific translocation domain in the effector protein. Efficient, T3SS-dependent translocation of purified YopH added in vitro was also observed when using coated Salmonella typhimurium strains, which implies that T3SS-mediated translocation of extracellular effector proteins is conserved among T3SS-dependent pathogens. Our results demonstrate that polarized T3SS-dependent translocation of proteins can be achieved through an intermediate extracellular step that can be reconstituted in vitro. These results indicate that translocation can occur by a different mechanism from the assumed single-step conduit model.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Membrana Celular/metabolismo , Citosol/metabolismo , Yersinia pseudotuberculosis/metabolismo , Proteínas da Membrana Bacteriana Externa/genética , Western Blotting , Cálcio/metabolismo , Membrana Celular/ultraestrutura , Citosol/microbiologia , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Microscopia Imunoeletrônica , Mutação , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Plasmídeos/genética , Transporte Proteico , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/metabolismo , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/fisiologia
11.
Microbiol Spectr ; 12(1): e0278123, 2024 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-38019016

RESUMO

IMPORTANCE: Unveiling gene co-expression networks in bacterial pathogens has the potential for gaining insights into their adaptive strategies within the host environment. Here, we developed Co-PATHOgenex, an interactive and user-friendly web application that enables users to construct networks from gene co-expressions using custom-defined thresholds (https://avicanlab.shinyapps.io/copathogenex/). The incorporated search functions and visualizations within the tool simplify the usage and facilitate the interpretation of the analysis output. Co-PATHOgenex also includes stress stimulons for various bacterial species, which can help identify gene products not previously associated with a particular stress condition.


Assuntos
Proteínas , Software , Redes Reguladoras de Genes , Bactérias/genética , RNA
12.
Nat Commun ; 15(1): 904, 2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38291037

RESUMO

Mast cells localize to mucosal tissues and contribute to innate immune defense against infection. How mast cells sense, differentiate between, and respond to bacterial pathogens remains a topic of ongoing debate. Using the prototype enteropathogen Salmonella Typhimurium (S.Tm) and other related enterobacteria, here we show that mast cells can regulate their cytokine secretion response to distinguish between extracellular and invasive bacterial infection. Tissue-invasive S.Tm and mast cells colocalize in the mouse gut during acute Salmonella infection. Toll-like Receptor 4 (TLR4) sensing of extracellular S.Tm, or pure lipopolysaccharide, causes a modest induction of cytokine transcripts and proteins, including IL-6, IL-13, and TNF. By contrast, type-III-secretion-system-1 (TTSS-1)-dependent S.Tm invasion of both mouse and human mast cells triggers rapid and potent inflammatory gene expression and >100-fold elevated cytokine secretion. The S.Tm TTSS-1 effectors SopB, SopE, and SopE2 here elicit a second activation signal, including Akt phosphorylation downstream of effector translocation, which combines with TLR activation to drive the full-blown mast cell response. Supernatants from S.Tm-infected mast cells boost macrophage survival and maturation from bone-marrow progenitors. Taken together, this study shows that mast cells can differentiate between extracellular and host-cell invasive enterobacteria via a two-step activation mechanism and tune their inflammatory output accordingly.


Assuntos
Infecções por Enterobacteriaceae , Infecções por Salmonella , Camundongos , Animais , Humanos , Mastócitos , Infecções por Salmonella/microbiologia , Salmonella typhimurium/genética , Citocinas/metabolismo
13.
Infect Immun ; 81(1): 11-22, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23090955

RESUMO

The enteropathogen Yersinia pseudotuberculosis can survive in the harsh environment of lymphoid compartments that abounds in immune cells. This capacity is dependent on the plasmid-encoded Yersinia outer proteins (Yops) that are delivered into the host cell via a mechanism involving the Yersinia type III secretion system. We show that the virulence protein YopK has a role in the mechanism by which Y. pseudotuberculosis avoids the polymorphonuclear leukocyte or neutrophil (PMN) defense. A yopK mutant, which is attenuated in the mouse infection model, where it fails to cause systemic infection, was found to colonize Peyer's patches and mesenteric lymph nodes more rapidly than the wild-type strain. Further, in mice lacking PMNs, the yopK mutant caused full disease with systemic spread and typical symptoms. Analyses of effects on PMNs revealed that both the wild-type strain and the yopK mutant inhibited internalization and reactive oxygen species production, as well as neutrophil extracellular trap formation by PMNs. However, the wild-type strain effectively avoided induction of PMN death, whereas the mutant caused a necrosis-like PMN death. Taken together, our results indicate that YopK is required for the ability of Yersinia to resist the PMN defense, which is critical for the virulence of the pathogen. We suggest a mechanism whereby YopK functions to prevent unintended Yop delivery and thereby PMN disruption, resulting in necrosis-like cell death, which would enhance the inflammatory response favoring the host.


Assuntos
Proteínas da Membrana Bacteriana Externa/imunologia , Neutrófilos/imunologia , Infecções por Yersinia pseudotuberculosis/imunologia , Yersinia pseudotuberculosis/imunologia , Animais , Morte Celular/imunologia , Feminino , Humanos , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Necrose/imunologia , Nódulos Linfáticos Agregados/imunologia , Espécies Reativas de Oxigênio/imunologia , Infecções por Yersinia pseudotuberculosis/sangue
14.
Microb Pathog ; 53(3-4): 154-61, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22771374

RESUMO

Dendritic cells play an important role in the immune response against pathogens, as they are responsible for the activation and control of both innate and adaptive immune responses. The CD11c-DTR(tg) model, which allows transient elimination of dendritic cells by diphtheria toxin-treatment (DTx), has been extensively used to study the importance of this immune cell during steady-state and infection conditions in mice. Infecting dendritic cell-depleted mice orally with Yersinia pseudotuberculosis results in a markedly reduced level of infection compared with infection of non-depleted mice. We show here that it is not the lack of dendritic cells per se that is responsible for the reduced infection efficiency, instead it is an immune response induced by the DTx-treatment that prevents the bacteria from establishing colonization in Peyer's patches. The DTx-induced depletion initiates an immune response, with elevated serum levels of keratinocyte-derived cytokine (KC) and recruitment of polymorphonuclear neutrophils to dendritic cell-containing organs, such as Peyer's patches. Since the window for having an animal depleted of dendritic cells is limited in time for this model, the DTx-mediated effect on the immune system complicates the use of this model in studies of early events during bacterial infections.


Assuntos
Antígenos CD11/genética , Toxina Diftérica/imunologia , Gastroenteropatias/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Infecções por Yersinia pseudotuberculosis/imunologia , Yersinia pseudotuberculosis/fisiologia , Animais , Antígenos CD11/imunologia , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Toxina Diftérica/genética , Modelos Animais de Doenças , Feminino , Gastroenteropatias/genética , Gastroenteropatias/microbiologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Humanos , Sistema Imunitário , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/microbiologia , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/imunologia , Infecções por Yersinia pseudotuberculosis/genética , Infecções por Yersinia pseudotuberculosis/microbiologia
15.
Nat Commun ; 12(1): 3282, 2021 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-34078900

RESUMO

Bacterial processes necessary for adaption to stressful host environments are potential targets for new antimicrobials. Here, we report large-scale transcriptomic analyses of 32 human bacterial pathogens grown under 11 stress conditions mimicking human host environments. The potential relevance of the in vitro stress conditions and responses is supported by comparisons with available in vivo transcriptomes of clinically important pathogens. Calculation of a probability score enables comparative cross-microbial analyses of the stress responses, revealing common and unique regulatory responses to different stresses, as well as overlapping processes participating in different stress responses. We identify conserved and species-specific 'universal stress responders', that is, genes showing altered expression in multiple stress conditions. Non-coding RNAs are involved in a substantial proportion of the responses. The data are collected in a freely available, interactive online resource (PATHOgenex).


Assuntos
Regulação Bacteriana da Expressão Gênica , Bactérias Gram-Negativas/genética , Bactérias Gram-Positivas/genética , RNA Bacteriano/genética , Estresse Fisiológico/genética , Transcriptoma , Adaptação Fisiológica/genética , Atlas como Assunto , Bases de Dados Genéticas , Perfilação da Expressão Gênica , Genes Bacterianos , Bactérias Gram-Negativas/classificação , Bactérias Gram-Negativas/metabolismo , Bactérias Gram-Negativas/patogenicidade , Bactérias Gram-Positivas/classificação , Bactérias Gram-Positivas/metabolismo , Bactérias Gram-Positivas/patogenicidade , Interações entre Hospedeiro e Microrganismos/genética , Humanos , Internet , Microbiota/genética , Filogenia , RNA Bacteriano/metabolismo
16.
Mol Oncol ; 15(4): 968-986, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33275817

RESUMO

Selective ERα modulator, tamoxifen, is well tolerated in a heavily pretreated castration-resistant prostate cancer (PCa) patient cohort. However, its targeted gene network and whether expression of intratumor ERα due to androgen deprivation therapy (ADT) may play a role in PCa progression is unknown. In this study, we examined the inhibitory effect of tamoxifen on castration-resistant PCa in vitro and in vivo. We found that tamoxifen is a potent compound that induced a high degree of apoptosis and significantly suppressed growth of xenograft tumors in mice, at a degree comparable to ISA-2011B, an inhibitor of PIP5K1α that acts upstream of PI3K/AKT survival signaling pathway. Moreover, depletion of tumor-associated macrophages using clodronate in combination with tamoxifen increased inhibitory effect of tamoxifen on aggressive prostate tumors. We showed that both tamoxifen and ISA-2011B exert their on-target effects on prostate cancer cells by targeting cyclin D1 and PIP5K1α/AKT network and the interlinked estrogen signaling. Combination treatment using tamoxifen together with ISA-2011B resulted in tumor regression and had superior inhibitory effect compared with that of tamoxifen or ISA-2011B alone. We have identified sets of genes that are specifically targeted by tamoxifen, ISA-2011B or combination of both agents by RNA-seq. We discovered that alterations in unique gene signatures, in particular estrogen-related marker genes are associated with poor patient disease-free survival. We further showed that ERα interacted with PIP5K1α through formation of protein complexes in the nucleus, suggesting a functional link. Our finding is the first to suggest a new therapeutic potential to inhibit or utilize the mechanisms related to ERα, PIP5K1α/AKT network, and MMP9/VEGF signaling axis, providing a strategy to treat castration-resistant ER-positive subtype of prostate cancer tumors with metastatic potential.


Assuntos
Dicetopiperazinas/uso terapêutico , Receptor alfa de Estrogênio/antagonistas & inibidores , Indóis/uso terapêutico , Isoquinolinas/uso terapêutico , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Tamoxifeno/uso terapêutico , Animais , Apoptose , Humanos , Masculino , Camundongos , Camundongos Nus , Células PC-3 , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt , RNA-Seq , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Front Microbiol ; 12: 706846, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34408737

RESUMO

The treatment of invasive Escherichia coli infections is a challenge because of the emergence and rapid spread of multidrug resistant strains. Particular problems are those strains that produce extended spectrum ß-lactamases (ESBL's). Although the global characterization of these enzymes is advanced, knowledge of their molecular basis among clinical E. coli isolates in Ethiopia is extremely limited. This study intends to address this knowledge gap. The study combines antimicrobial resistance profiling and molecular epidemiology of ESBL genes among 204 E. coli clinical isolates collected from patient urine, blood, and pus at four geographically distinct health facilities in Ethiopia. All isolates exhibited multidrug resistance, with extensive resistance to ampicillin and first to fourth line generation cephalosporins and sulfamethoxazole-trimethoprim and ciprofloxacin. Extended spectrum ß-lactamase genes were detected in 189 strains, and all but one were positive for CTX-Ms ß-lactamases. Genes encoding for the group-1 CTX-Ms enzymes were most prolific, and CTX-M-15 was the most common ESBL identified. Group-9 CTX-Ms including CTX-M-14 and CTX-27 were detected only in 12 isolates and SHV ESBL types were identified in just 8 isolates. Bacterial typing revealed a high amount of strains associated with the B2 phylogenetic group. Crucially, the international high risk clones ST131 and ST410 were among the sequence types identified. This first time study revealed a high prevalence of CTX-M type ESBL's circulating among E. coli clinical isolates in Ethiopia. Critically, they are associated with multidrug resistance phenotypes and high-risk clones first characterized in other parts of the world.

18.
Cell Microbiol ; 11(12): 1750-67, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19681909

RESUMO

One important feature of Yersinia pseudotuberculosis that enables resistance against the host immune defence is delivery of the antiphagocytic effectors YopH and YopE into phagocytic cells. The tyrosine phosphatase YopH influences integrin signalling, and YopE impairs cytoskeletal dynamics by inactivating Rho GTPases. Here, we report the impact of these effectors on internalization by dendritic cells (DCs), which internalize antigens to orchestrate host immune responses. We found that this pathogen resists internalization by DCs via YopE. YopH that is important for blocking phagocytosis by macrophages and neutrophils and which is also present inside the DCs does not contribute to the resistance. However, the YopH targets Fyb and p130Cas show higher expression levels in macrophages than in DCs. Furthermore, live cell microscopy revealed that the cells internalize Y. pseudotuberculosis in different ways: the macrophages utilize a locally restricted receptor-mediated zipper mechanism, whereas DCs utilize macropinocytosis involving constitutive ruffling that randomly catches bacteria into membrane folds. We conclude that YopH impacts early phagocytic signalling from the integrin receptor to which the bacterium binds and that this tight receptor-mediated stimulation is absent in DC macropinocytosis. Inactivation of cytoskeletal dynamics by YopE affects ruffling activity and hence also internalization. The different modes of internalization can be coupled to the major functions of these respective cell types: elimination by phagocytosis and antigen sampling.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Células Dendríticas/metabolismo , Macrófagos/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Fatores de Virulência/metabolismo , Infecções por Yersinia pseudotuberculosis/imunologia , Infecções por Yersinia pseudotuberculosis/metabolismo , Yersinia pseudotuberculosis/fisiologia , Animais , Proteínas da Membrana Bacteriana Externa/imunologia , Técnicas de Cultura de Células , Linhagem Celular , Células Dendríticas/citologia , Células Dendríticas/imunologia , Feminino , Interações Hospedeiro-Patógeno , Humanos , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Fagocitose , Proteínas Tirosina Fosfatases/imunologia , Virulência , Fatores de Virulência/imunologia , Yersinia pseudotuberculosis/citologia , Yersinia pseudotuberculosis/patogenicidade , Infecções por Yersinia pseudotuberculosis/microbiologia
19.
mSystems ; 5(6)2020 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-33172972

RESUMO

RpoN, an alternative sigma factor commonly known as σ54, is implicated in persistent stages of Yersinia pseudotuberculosis infections in which genes associated with this regulator are upregulated. We here combined phenotypic and genomic assays to provide insight into its role and function in this pathogen. RpoN was found essential for Y. pseudotuberculosis virulence in mice, and in vitro functional assays showed that it controls biofilm formation and motility. Mapping genome-wide associations of Y. pseudotuberculosis RpoN using chromatin immunoprecipitation coupled with next-generation sequencing identified an RpoN binding motif located at 103 inter- and intragenic sites on both sense and antisense strands. Deletion of rpoN had a large impact on gene expression, including downregulation of genes encoding proteins involved in flagellar assembly, chemotaxis, and quorum sensing. There were also clear indications of cross talk with other sigma factors, together with indirect effects due to altered expression of other regulators. Matching differential gene expression with locations of the binding sites implicated around 130 genes or operons potentially activated or repressed by RpoN. Mutagenesis of selected intergenic binding sites confirmed both positive and negative regulatory effects of RpoN binding. Corresponding mutations of intragenic sense sites had less impact on associated gene expression. Surprisingly, mutating intragenic sites on the antisense strand commonly reduced expression of genes carried by the corresponding sense strand.IMPORTANCE The alternative sigma factor RpoN (σ54), which is widely distributed in eubacteria, has been implicated in controlling gene expression of importance for numerous functions including virulence. Proper responses to host environments are crucial for bacteria to establish infection, and regulatory mechanisms involved are therefore of high interest for development of future therapeutics. Little is known about the function of RpoN in the intestinal pathogen Y. pseudotuberculosis, and we therefore investigated its regulatory role in this pathogen. This regulator was indeed found to be critical for establishment of infection in mice, likely involving its requirement for motility and biofilm formation. The RpoN regulon involved both activating and suppressive effects on gene expression which could be confirmed with mutagenesis of identified binding sites. This is the first study of its kind of RpoN in Y. pseudotuberculosis, revealing complex regulation of gene expression involving both productive and silent effects of its binding to DNA, providing important information about RpoN regulation in enterobacteria.

20.
J Proteomics ; 195: 33-40, 2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30641234

RESUMO

Campylobacter jejuni is the major cause of bacterial gastroenteritis in humans. In contrast, colonization in avian hosts is asymptomatic. Body temperature differs between human (37 °C) and avian (42 °C) hosts, and bacterial growth in 37 °C is therefore a potential cue for higher virulence properties during human infection. The proteome of the bacteria was previously shown to be altered by temperature. Here we investigated whether temperature has an effect on the C. jejuni outer membrane vesicle (OMV) proteome, as OMVs are considered to be bacterial vehicles for protein delivery and might play a role during infection. OMVs isolated from C. jejuni strain 81-176 grown at 37 °C and 42 °C were analyzed by LC-ESI-MS/MS. 181 proteins were detected in both sample groups, one protein was exclusively present, and three were absent in OMVs from 37 °C. Of the 181 proteins, 59 were differentially expressed; 30 proteins were detected with higher abundance, and 29 proteins with lower abundance at 37 °C. Among the more highly abundant proteins, significantly more proteins were predicted to be associated with virulence. These data show that temperature has an impact on the property of the OMVs, and this might affect the outcome of colonization/infection by C. jejuni in different hosts. SIGNIFICANCE: While C. jejuni is considered as a commensal bacterium in avian hosts, it causes symptomatic infection in humans. As the host body temperature is one differentiating factor, the growth temperature can potentially affect the virulence properties of the bacteria, but also OMVs that are released during growth. By using a proteomic approach, in this study, we observed that the protein content of OMVs isolated from C. jejuni is affected by growth temperature and that more proteins related to virulence are associated with OMVs at 37°C growth temperature. This property indicates that C. jejuni OMVs have a potential role for the outcome of human infection.


Assuntos
Proteínas da Membrana Bacteriana Externa/biossíntese , Temperatura Corporal , Campylobacter jejuni , Fatores de Virulência/biossíntese , Campylobacter jejuni/crescimento & desenvolvimento , Campylobacter jejuni/patogenicidade , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA