Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Vet Res ; 55(1): 81, 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38926765

RESUMO

The escalation of antibiotic resistance, pandemics, and nosocomial infections underscores the importance of research in both animal and human infectious diseases. Recent advancements in three-dimensional tissue cultures, or "organoids", have revolutionized the development of in vitro models for infectious diseases. Our study conducts a bibliometric analysis on the use of organoids in modeling infectious diseases, offering an in-depth overview of this field's current landscape. We examined scientific contributions from 2009 onward that focused on organoids in host‒pathogen interactions using the Web of Science Core Collection and OpenAlex database. Our analysis included temporal trends, reference aging, author, and institutional productivity, collaborative networks, citation metrics, keyword cluster dynamics, and disruptiveness of organoid models. VOSviewer, CiteSpace, and Python facilitated this analytical assessment. The findings reveal significant growth and advancements in organoid-based infectious disease research. Analysis of keywords and impactful publications identified three distinct developmental phases in this area that were significantly influenced by outbreaks of Zika and SARS-CoV-2 viruses. The research also highlights the synergistic efforts between academia and publishers in tackling global pandemic challenges. Through mostly consolidating research efforts, organoids are proving to be a promising tool in infectious disease research for both human and animal infectious disease. Their integration into the field necessitates methodological refinements for better physiological emulation and the establishment of extensive organoid biobanks. These improvements are crucial for fully harnessing the potential of organoids in understanding infectious diseases and advancing the development of targeted treatments and vaccines.


Assuntos
Bibliometria , Organoides , Organoides/virologia , Animais , Humanos , Doenças Transmissíveis/veterinária , Doenças Transmissíveis/epidemiologia , Modelos Animais de Doenças , COVID-19/epidemiologia , COVID-19/virologia
2.
Vet Res ; 54(1): 63, 2023 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-37525204

RESUMO

Chicken infection with Salmonella Typhimurium is an important source of foodborne human diseases. Salmonella colonizes the avian intestinal tract and more particularly the caecum, without causing symptoms. This thus poses a challenge for the prevention of foodborne transmission. Until now, studies on the interaction of Salmonella with the avian gut intestine have been limited by the absence of in vitro intestinal culture models. Here, we established intestinal crypt-derived chicken organoids to better decipher the impact of Salmonella intracellular replication on avian intestinal epithelium. Using a 3D organoid model, we observed a significantly higher replication rate of the intracellular bacteria in caecal organoids than in ileal organoids. Our model thus recreates intracellular environment, allowing Salmonella replication of avian epithelium according to the intestinal segment. Moreover, an inhibition of the cellular proliferation was observed in infected ileal and caecal organoids compared to uninfected organoids. This appears with a higher effect in ileal organoids, as well as a higher cytokine and signaling molecule response in infected ileal organoids at 3 h post-infection (hpi) than in caecal organoids that could explain the lower replication rate of Salmonella observed later at 24 hpi. To conclude, this study demonstrates that the 3D organoid is a model allowing to decipher the intracellular impact of Salmonella on the intestinal epithelium cell response and illustrates the importance of the gut segment used to purify stem cells and derive organoids to specifically study epithelial cell -Salmonella interaction.


Assuntos
Galinhas , Salmonella typhimurium , Humanos , Animais , Salmonella typhimurium/fisiologia , Intestinos , Mucosa Intestinal/microbiologia , Ceco , Organoides/microbiologia
3.
BMC Microbiol ; 21(1): 153, 2021 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-34020586

RESUMO

BACKGROUND: Salmonella can invade host cells via a type three secretion system called T3SS-1 and its outer membrane proteins, PagN and Rck. However, the mechanism of PagN-dependent invasion pathway used by Salmonella enterica, subspecies enterica serovar Typhimurium remains unclear. RESULTS: Here, we report that PagN is well conserved and widely distributed among the different species and subspecies of Salmonella. We showed that PagN of S. Typhimurium was sufficient and necessary to enable non-invasive E. coli over-expressing PagN and PagN-coated beads to bind to and invade different non-phagocytic cells. According to the literature, PagN is likely to interact with heparan sulfate proteoglycan (HSPG) as PagN-mediated invasion could be inhibited by heparin treatment in a dose-dependent manner. This report shows that this interaction is not sufficient to allow the internalization mechanism. Investigation of the role of ß1 integrin as co-receptor showed that mouse embryo fibroblasts genetically deficient in ß1 integrin were less permissive to PagN-mediated internalization. Moreover, PagN-mediated internalization was fully inhibited in glycosylation-deficient pgsA-745 cells treated with anti-ß1 integrin antibody, supporting the hypothesis that ß1 integrin and HSPG cooperate to induce the PagN-mediated internalization mechanism. In addition, use of specific inhibitors and expression of dominant-negative derivatives demonstrated that tyrosine phosphorylation and class I phosphatidylinositol 3-kinase were crucial to trigger PagN-dependent internalization, as for the Rck internalization mechanism. Finally, scanning electron microscopy with infected cells showed microvillus-like extensions characteristic of Zipper-like structure, engulfing PagN-coated beads and E. coli expressing PagN, as observed during Rck-mediated internalization. CONCLUSIONS: Our results supply new comprehensions into T3SS-1-independent invasion mechanisms of S. Typhimurium and highly indicate that PagN induces a phosphatidylinositol 3-kinase signaling pathway, leading to a Zipper-like entry mechanism as the Salmonella outer membrane protein Rck.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Infecções por Salmonella/microbiologia , Salmonella typhimurium/metabolismo , Animais , Proteínas da Membrana Bacteriana Externa/genética , Linhagem Celular , Fibroblastos/metabolismo , Fibroblastos/microbiologia , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Camundongos , Infecções por Salmonella/genética , Infecções por Salmonella/metabolismo , Salmonella typhimurium/genética , Sistemas de Secreção Tipo III/genética , Sistemas de Secreção Tipo III/metabolismo
4.
Vet Res ; 52(1): 33, 2021 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-33632315

RESUMO

In livestock species, the monolayer of epithelial cells covering the digestive mucosa plays an essential role for nutrition and gut barrier function. However, research on farm animal intestinal epithelium has been hampered by the lack of appropriate in vitro models. Over the past decade, methods to culture livestock intestinal organoids have been developed in pig, bovine, rabbit, horse, sheep and chicken. Gut organoids from farm animals are obtained by seeding tissue-derived intestinal epithelial stem cells in a 3-dimensional culture environment reproducing in vitro the stem cell niche. These organoids can be generated rapidly within days and are formed by a monolayer of polarized epithelial cells containing the diverse differentiated epithelial progeny, recapitulating the original structure and function of the native epithelium. The phenotype of intestinal organoids is stable in long-term culture and reflects characteristics of the digestive segment of origin. Farm animal intestinal organoids can be amplified in vitro, cryopreserved and used for multiple experiments, allowing an efficient reduction of the use of live animals for experimentation. Most of the studies using livestock intestinal organoids were used to investigate host-microbe interactions at the epithelial surface, mainly focused on enteric infections with viruses, bacteria or parasites. Numerous other applications of farm animal intestinal organoids include studies on nutrient absorption, genome editing and bioactive compounds screening relevant for agricultural, veterinary and biomedical sciences. Further improvements of the methods used to culture intestinal organoids from farm animals are required to replicate more closely the intestinal tissue complexity, including the presence of non-epithelial cell types and of the gut microbiota. Harmonization of the methods used to culture livestock intestinal organoids will also be required to increase the reproducibility of the results obtained in these models. In this review, we summarize the methods used to generate and cryopreserve intestinal organoids in farm animals, present their phenotypes and discuss current and future applications of this innovative culture system of the digestive epithelium.


Assuntos
Animais Domésticos/anatomia & histologia , Técnicas de Cultura de Células/veterinária , Criopreservação/veterinária , Intestino Grosso/citologia , Intestino Delgado/citologia , Organoides/citologia , Animais , Técnicas de Cultura de Células/métodos , Criopreservação/métodos , Células Epiteliais/citologia , Mucosa Intestinal/citologia
5.
FASEB J ; 30(12): 4180-4191, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27609774

RESUMO

The Salmonella Rck outer membrane protein binds to the cell surface, which leads to bacterial internalization via a Zipper mechanism. This invasion process requires induction of cellular signals, including phosphorylation of tyrosine proteins, and activation of c-Src and PI3K, which arises as a result of an interaction with a host cell surface receptor. In this study, epidermal growth factor receptor (EGFR) was identified as the cell signaling receptor required for Rck-mediated adhesion and internalization. First, Rck-mediated adhesion and internalization were shown to be altered when EGFR expression and activity were modulated. Then, immunoprecipitations were performed to demonstrate the Rck-EGFR interaction. Furthermore, surface plasmon resonance biosensor and homogeneous time-resolved fluorescence technologies were used to demonstrate the direct interaction of Rck with the extracellular domain of human EGFR. Finally, our study strongly suggests a noncompetitive binding of Rck and EGF to EGFR. Overall, these results demonstrate that Rck is able to bind to EGFR and thereby establish a tight adherence to provide a signaling cascade, which leads to internalization of Rck-expressing bacteria.-Wiedemann, A., Mijouin, L., Ayoub, M. A., Barilleau, E., Canepa, S., Teixeira-Gomes, A. P., Le Vern, Y., Rosselin, M., Reiter, E., Velge, P. Identification of the epidermal growth factor receptor as the receptor for Salmonella Rck-dependent invasion.


Assuntos
Membrana Celular/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Salmonella/metabolismo , Proteína Tirosina Quinase CSK , Linhagem Celular , Escherichia coli , Fosforilação , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/fisiologia , Quinases da Família src/metabolismo
6.
Mol Microbiol ; 94(2): 254-71, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25080967

RESUMO

One important step for the pathogenesis of Salmonella is its ability to penetrate host cells. Recently, a new entry system involving the outer membrane protein Rck has been characterized. Previous studies have shown that the pefI-srgC locus, which contains rck, was regulated by the temperature and SdiA, the transcriptional regulator of quorum sensing in Salmonella. To decipher the regulation of rck by SdiA, we first confirmed the operon organization of the pefI-srgC locus. Using plasmid-based transcriptional fusions, we showed that only the predicted distal promoter upstream of pefI, PefIP2, displays an SdiA- and acyl-homoserine lactones-dependent activity while the predicted proximal PefIP1 promoter exhibits a very low activity independent on SdiA in our culture conditions. A direct and specific interaction of SdiA with this PefIP2 region was identified using electrophoretic mobility shift assays and surface plasmon resonance studies. We also observed that Rck expression is negatively regulated by the nucleoid-associated H-NS protein at both 25°C and 37°C. This work is the first demonstration of a direct regulation of genes by SdiA in Salmonella and will help further studies designed to identify environmental conditions required for Rck expression and consequently contribute to better characterize the role of this invasin in vivo.


Assuntos
Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Óperon , Salmonella typhimurium/genética , Transativadores/metabolismo , Fatores de Virulência/biossíntese , Fusão Gênica Artificial , DNA Bacteriano/metabolismo , Proteínas de Ligação a DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Perfilação da Expressão Gênica , Ordem dos Genes , Genes Bacterianos , Genes Reporter , Regiões Promotoras Genéticas , Ligação Proteica , Percepção de Quorum , Salmonella typhimurium/fisiologia , Ressonância de Plasmônio de Superfície , Temperatura , Fatores de Virulência/genética
7.
Vet Res ; 45: 81, 2014 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-25175996

RESUMO

Salmonella Gallinarum and Salmonella Enteritidis are genetically closely related however associated with different pathologies. Several studies have suggested that S. Gallinarum is less invasive in vitro than S. Enteritidis. In this study we confirm that the S. Gallinarum strains tested were much less invasive than the S. Enteritidis strains tested in cells of avian or human origin. In addition, the S. Gallinarum T3SS-1-dependent ability to invade host cells was delayed by two to three hours compared to S. Enteritidis, indicating that T3SS-1-dependent entry is less efficient in S. Gallinarum than S. Enteritidis. This was neither due to a decreased transcription of T3SS-1 related genes when bacteria come into contact with cells, as transcription of hilA, invF and sipA was similar to that observed for S. Enteritidis, nor to a lack of functionality of the S. Gallinarum T3SS-1 apparatus as this apparatus was able to secrete and translocate effector proteins into host cells. In contrast, genome comparison of four S. Gallinarum and two S. Enteritidis strains revealed that all S. Gallinarum genomes displayed the same point mutations in each of the main T3SS-1 effector genes sipA, sopE, sopE2, sopD and sopA.


Assuntos
Doenças das Aves Domésticas/microbiologia , Salmonelose Animal/microbiologia , Infecções por Salmonella/microbiologia , Salmonella enterica/fisiologia , Salmonella enterica/patogenicidade , Salmonella enteritidis/fisiologia , Salmonella enteritidis/patogenicidade , Animais , Aderência Bacteriana , Linhagem Celular , Linhagem Celular Tumoral , Galinhas , Humanos , Salmonella enterica/genética , Salmonella enteritidis/genética
8.
Virulence ; 15(1): 2357670, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38804638

RESUMO

Salmonella enterica subspecies enterica serovar Typhimurium is an intracellular pathogen that invades and colonizes the intestinal epithelium. Following bacterial invasion, Salmonella is enclosed within a membrane-bound vacuole known as a Salmonella-containing vacuole (SCV). However, a subset of Salmonella has the capability to prematurely rupture the SCV and escape, resulting in Salmonella hyper-replication within the cytosol of epithelial cells. A recently published RNA-seq study provides an overview of cytosolic and vacuolar upregulated genes and highlights pagN vacuolar upregulation. Here, using transcription kinetics, protein production profile, and immunofluorescence microscopy, we showed that PagN is exclusively produced by Salmonella in SCV. Gentamicin protection and chloroquine resistance assays were performed to demonstrate that deletion of pagN affects Salmonella replication by affecting the cytosolic bacterial population. This study presents the first example of a Salmonella virulence factor expressed within the endocytic compartment, which has a significant impact on the dynamics of Salmonella cytosolic hyper-replication.


Assuntos
Proteínas de Bactérias , Citosol , Salmonella typhimurium , Vacúolos , Fatores de Virulência , Salmonella typhimurium/genética , Salmonella typhimurium/patogenicidade , Citosol/microbiologia , Vacúolos/microbiologia , Vacúolos/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Humanos , Virulência , Infecções por Salmonella/microbiologia , Células HeLa , Células Epiteliais/microbiologia , Regulação Bacteriana da Expressão Gênica
9.
Sci Rep ; 14(1): 15160, 2024 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-38956132

RESUMO

In order to survive and replicate, Salmonella has evolved mechanisms to gain access to intestinal epithelial cells of the crypt. However, the impact of Salmonella Typhimurium on stem cells and progenitors, which are responsible for the ability of the intestinal epithelium to renew and protect itself, remains unclear. Given that intestinal organoids growth is sustained by stem cells and progenitors activity, we have used this model to document the effects of Salmonella Typhimurium infection on epithelial proliferation and differentiation, and compared it to an in vivo model of Salmonella infection in mice. Among gut segments, the caecum was preferentially targeted by Salmonella. Analysis of infected crypts and organoids demonstrated increased length and size, respectively. mRNA transcription profiles of infected crypts and organoids pointed to upregulated EGFR-dependent signals, associated with a decrease in secretory cell lineage differentiation. To conclude, we show that organoids are suited to mimic the impact of Salmonella on stem cells and progenitors cells, carrying a great potential to drastically reduce the use of animals for scientific studies on that topic. In both models, the EGFR pathway, crucial to stem cells and progenitors proliferation and differentiation, is dysregulated by Salmonella, suggesting that repeated infections might have consequences on crypt integrity and further oncogenesis.


Assuntos
Diferenciação Celular , Receptores ErbB , Organoides , Infecções por Salmonella , Salmonella typhimurium , Células-Tronco , Animais , Organoides/microbiologia , Células-Tronco/metabolismo , Camundongos , Salmonella typhimurium/patogenicidade , Salmonella typhimurium/fisiologia , Infecções por Salmonella/microbiologia , Infecções por Salmonella/patologia , Receptores ErbB/metabolismo , Receptores ErbB/genética , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Proliferação de Células , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL
10.
J Biol Chem ; 287(37): 31148-54, 2012 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-22810232

RESUMO

The Salmonella outer membrane protein Rck mediates a Zipper entry mechanism controlled by tyrosine phosphorylation and class I phosphatidylinositol 3-kinase (PI 3-kinase). However, the underlying mechanism leading to this signaling cascade remains unclear. The present study showed that using Rck-coated beads or Rck-overexpressing Escherichia coli, Rck-mediated actin polymerization and invasion were blocked by PP2, a Src family tyrosine kinase inhibitor. In addition, phosphorylation of Src family kinases significantly increased after stimulation with Rck. The specific contribution of c-Src, one member of the Src family kinases, was demonstrated using c-Src-deficient fibroblasts or c-Src siRNA transfected epithelial cells. We also observed that Rck-mediated internalization led to the formation of a complex between c-Src and at least one tyrosine-phosphorylated protein. Furthermore, our results revealed that the c-Src signal molecule was upstream of PI 3-kinase during the Rck-mediated signaling pathway as Rck-mediated PI 3-kinase activation was blocked by PP2, and PI 3-kinase inhibitor had no effect on the Src phosphorylation. These results demonstrate the involvement of c-Src upstream of the PI 3-kinase in the Zipper entry process mediated by Rck.


Assuntos
Proteínas de Bactérias/metabolismo , Complexos Multiproteicos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Infecções por Salmonella/metabolismo , Salmonella enteritidis/metabolismo , Proteínas de Bactérias/genética , Proteína Tirosina Quinase CSK , Linhagem Celular , Ativação Enzimática/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Complexos Multiproteicos/genética , Fosfatidilinositol 3-Quinases/genética , Proteínas Tirosina Quinases/genética , Infecções por Salmonella/genética , Salmonella enteritidis/genética , Quinases da Família src
11.
FASEB J ; 26(4): 1569-81, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22210834

RESUMO

The Salmonella outer membrane protein Rck mediates a Zipper-like entry mechanism controlled by Rac, the Arp2/3 complex, and actin polymerization. However, little is known about the early steps leading to Rac activation and Rck-mediated internalization. The use of pharmacological inhibitors or PI 3-kinase dominant-negative mutant induced more than 80% less invasion without affecting attachment. Moreover, Rck-mediated internalization caused an increase in the association of p85 with at least one tyrosine-phosphorylated protein, indicating that class I PI 3-kinase activity was stimulated. We also report that this PI 3-kinase activity is essential for Rac1 activation. However, Rac recruitment at the Rck-mediated entry site was independent of its activation. Using a pharmacological approach or Akt-knockout cells, we also demonstrated that Akt was phosphorylated in response to Rck-mediated internalization as demonstrated by immunoblotting analysis and that all three Akt isoforms were required during this process. Overall, our results describe a signaling pathway involving tyrosine phosphorylation, class I PI 3-kinase, Akt activation, and Rac activation, leading to Rck-dependent Zipper entry. The specificity of this signaling pathway with regard to that of the type 3 secretion system, which is the other invasion process of Salmonella, is discussed.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Salmonella enteritidis/metabolismo , Salmonella enteritidis/patogenicidade , Transdução de Sinais/fisiologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Proteínas da Membrana Bacteriana Externa/genética , Linhagem Celular , Células Cultivadas , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Camundongos , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Salmonella enteritidis/citologia , Proteínas rac1 de Ligação ao GTP/genética
12.
Front Microbiol ; 13: 906238, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35733975

RESUMO

Salmonella enterica serovars are invasive gram-negative bacteria, causing a wide range of diseases from gastroenteritis to typhoid fever, representing a public health threat around the world. Salmonella gains access to the intestinal lumen after oral ingestion of contaminated food or water. The crucial initial step to establish infection is the interaction with the intestinal epithelium. Human-adapted serovars such as S. Typhi or S. Paratyphi disseminate to systemic organs and induce life-threatening disease known as typhoid fever, whereas broad-host serovars such as S. Typhimurium usually are limited to the intestine and responsible for gastroenteritis in humans. To overcome intestinal epithelial barrier, Salmonella developed mechanisms to induce cellular invasion, intracellular replication and to face host defence mechanisms. Depending on the serovar and the respective host organism, disease symptoms differ and are linked to the ability of the bacteria to manipulate the epithelial barrier for its own profit and cross the intestinal epithelium. This review will focus on S. Typhimurium (STm). To better understand STm pathogenesis, it is crucial to characterize the crosstalk between STm and the intestinal epithelium and decipher the mechanisms and epithelial cell types involved. Thus, the purpose of this review is to summarize our current knowledge on the molecular dialogue between STm and the various cell types constituting the intestinal epithelium with a focus on the mechanisms developed by STm to cross the intestinal epithelium and access to subepithelial or systemic sites and survive host defense mechanisms.

13.
Microbiology (Reading) ; 157(Pt 3): 839-847, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21109565

RESUMO

Salmonella causes a wide range of diseases from acute gastroenteritis to systemic typhoid fever, depending on the host. To invade non-phagocytic cells, Salmonella has developed different mechanisms. The main invasion system requires a type III secretion system (T3SS) known as T3SS-1, which promotes a Trigger entry mechanism. However, other invasion factors have recently been described in Salmonella, including Rck and PagN, which were not expressed under our bacterial culture conditions. Based on these observations, we used adhesion and invasion assays to analyse the respective roles of Salmonella Enteritidis T3SS-1-dependent and -independent invasion processes at different times of infection. Diverse cell lines and cell types were tested, including endothelial, epithelial and fibroblast cells. We demonstrated that cell susceptibility to the T3SS-1-independent entry differs by a factor of nine between the most and the least permissive cell lines tested. In addition, using scanning electron and confocal microscopy, we showed that T3SS-1-independent entry into cells was characterized by a Trigger-like alteration, as for the T3SS-1-dependent entry, and also by Zipper-like cellular alteration. Our results demonstrate for what is believed to be the first time that Salmonella can induce Trigger-like entry independently of T3SS-1 and can induce Zipper-like entry independently of Rck. Overall, these data open new avenues for discovering new invasion mechanisms in Salmonella.


Assuntos
Proteínas de Bactérias/metabolismo , Células Endoteliais/microbiologia , Células Epiteliais/microbiologia , Fibroblastos/microbiologia , Especificidade de Hospedeiro , Salmonella enteritidis/patogenicidade , Células 3T3 , Actinas/metabolismo , Animais , Aderência Bacteriana/fisiologia , Sistemas de Secreção Bacterianos/fisiologia , Linhagem Celular , Membrana Celular/metabolismo , Membrana Celular/microbiologia , Células Endoteliais/metabolismo , Células Epiteliais/metabolismo , Fibroblastos/metabolismo , Células HT29 , Humanos , Camundongos , Microscopia Confocal , Salmonella enteritidis/metabolismo , Salmonella enteritidis/fisiologia
14.
J Cell Biol ; 172(7): 1069-79, 2006 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-16567504

RESUMO

AlphaMbeta2 integrins mediate phagocytosis of opsonized particles in a process controlled by RhoA, Rho kinase, myosin II, Arp2/3, and actin polymerization. AlphaMbeta2, Rho, Arp2/3, and F-actin accumulate underneath bound particles; however, the mechanism regulating Rho function during alphaMbeta2-mediated phagocytosis is poorly understood. We report that the binding of C3bi-opsonized sheep red blood cells (RBCs) to alphaMbeta2 increases Rho-GTP, but not Rac-GTP, levels. Deletion of the cytoplasmic domain of beta2, but not of alphaM, abolished Rho recruitment and activation, as well as phagocytic uptake. Interestingly, a 16-amino acid (aa) region in the membrane-proximal half of the beta2 cytoplasmic domain was necessary for activating Rho. Three COOH-terminal residues (aa 758-760) were essential for beta2-induced accumulation of Rho at complement receptor 3 (CR3) phagosomes. Activation of Rho was necessary, but not sufficient, for its stable recruitment underneath bound particles or for uptake. However, recruitment of active Rho was sufficient for phagocytosis. Our data shed light on the mechanism of outside-in signaling, from ligated integrins to the activation of Rho GTPase signaling.


Assuntos
Antígenos CD18/fisiologia , Fagocitose/fisiologia , Proteína rhoA de Ligação ao GTP/fisiologia , Actinas/metabolismo , Sequência de Aminoácidos , Animais , Antígenos CD18/genética , Antígenos CD18/metabolismo , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Complemento C3b/química , Complemento C3b/metabolismo , Eritrócitos/química , Eritrócitos/metabolismo , Guanosina Trifosfato/metabolismo , Antígeno de Macrófago 1/genética , Antígeno de Macrófago 1/metabolismo , Antígeno de Macrófago 1/fisiologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Dados de Sequência Molecular , Mutação , Fagocitose/efeitos dos fármacos , Fagossomos/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Treonina/genética , Transfecção , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
15.
J Exp Med ; 196(8): 1017-24, 2002 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-12391013

RESUMO

A characteristic of the three human-pathogenic Yersinia spp. (the plague agent Yersinia pestis and the enteropathogenic Yersinia pseudotuberculosis and Yersinia enterocolitica) is the expression of the virulence (V)-antigen (LcrV). LcrV is a released protein which is involved in contact-induced secretion of yersinia antihost proteins and in evasion of the host's innate immune response. Here we report that recombinant LcrV signals in a CD14- and toll-like receptor 2 (TLR2)-dependent fashion leading to immunosuppression by interleukin 10 induction. The impact of this immunosuppressive effect for yersinia pathogenesis is underlined by the observation that TLR2-deficient mice are less susceptible to oral Y. enterocolitica infection than isogenic wild-type animals. In summary, these data demonstrate a new ligand specificity of TLR2, as LcrV is the first known secreted and nonlipidated virulence-associated protein of a Gram-negative bacterium using TLR2 for cell activation. We conclude that yersiniae might exploit host innate pattern recognition molecules and defense mechanisms to evade the host immune response.


Assuntos
Antígenos de Bactérias/fisiologia , Proteínas de Drosophila , Terapia de Imunossupressão , Interleucina-10/fisiologia , Receptores de Lipopolissacarídeos/fisiologia , Glicoproteínas de Membrana/fisiologia , Receptores de Superfície Celular/fisiologia , Yersinia/fisiologia , Sequência de Aminoácidos , Animais , Antígenos de Bactérias/química , Antígenos de Bactérias/metabolismo , Interleucina-10/biossíntese , Lipopolissacarídeos/metabolismo , Macrófagos Peritoneais/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Proteínas Citotóxicas Formadoras de Poros , Transdução de Sinais/fisiologia , Receptor 2 Toll-Like , Receptores Toll-Like , Yersinia/imunologia , Yersiniose/imunologia
16.
Mol Biol Cell ; 18(3): 976-85, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17202407

RESUMO

The cytoskeletal, actin-binding protein talin has been previously implicated in phagocytosis in Dictyostelium discoideum and mammalian phagocytes. However, its mechanism of action during internalization is not understood. Our data confirm that endogenous talin can occasionally be found at phagosomes forming around IgG- and C3bi-opsonized red blood cells in macrophages. Remarkably, talin knockdown specifically abrogates uptake through complement receptor 3 (CR3, CD11b/CD18, alpha(M)beta(2) integrin) and not through the Fc gamma receptor. We show that talin physically interacts with CR3/alpha(M)beta(2) and that this interaction involves the talin head domain and residues W747 and F754 in the beta(2) integrin cytoplasmic domain. The CR3/alpha(M)beta(2)-talin head interaction controls not only talin recruitment to forming phagosomes but also CR3/alpha(M)beta(2) binding activity, both in macrophages and transfected fibroblasts. However, the talin head domain alone cannot support phagocytosis. Our results establish for the first time at least two distinct roles for talin during CR3/alpha(M)beta(2)-mediated phagocytosis, most noticeably activation of the CR3/alpha(M)beta(2) receptor and phagocytic uptake.


Assuntos
Antígeno de Macrófago 1/metabolismo , Fagocitose , Talina/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Eritrócitos/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imunoglobulina G/imunologia , Antígeno de Macrófago 1/química , Camundongos , Dados de Sequência Molecular , Fenilalanina , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Receptores de IgG/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Ovinos , Triptofano
17.
Front Cell Infect Microbiol ; 10: 586934, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33330131

RESUMO

Salmonella Typhimurium expresses on its outer membrane the protein Rck which interacts with the epidermal growth factor receptor (EGFR) of the plasma membrane of the targeted host cells. This interaction activates signaling pathways, leading to the internalization of Salmonella. Since EGFR plays a key role in cell proliferation, we sought to determine the influence of Rck mediated infection on the host cell cycle. By analyzing the DNA content of uninfected and infected cells using flow cytometry, we showed that the Rck-mediated infection induced a delay in the S-phase (DNA replication phase) of the host cell cycle, independently of bacterial internalization. We also established that this Rck-dependent delay in cell cycle progression was accompanied by an increased level of host DNA double strand breaks and activation of the DNA damage response. Finally, we demonstrated that the S-phase environment facilitated Rck-mediated bacterial internalization. Consequently, our results suggest that Rck can be considered as a cyclomodulin with a genotoxic activity.


Assuntos
Proteínas da Membrana Bacteriana Externa , Salmonella typhimurium , Proteínas da Membrana Bacteriana Externa/genética , Proteínas de Bactérias , Divisão Celular , Membrana Celular , Salmonella typhimurium/genética , Transdução de Sinais
18.
Front Microbiol ; 10: 541, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30972041

RESUMO

Defensins are natural antimicrobial peptides. The avian beta-defensin AvBD7 isolated from the chicken bone marrow possess broad antibacterial spectrum and strong resistance to proteolysis. However, its ability to fight systemic infections of major concern for public health, such as salmonellosis, is unknown. As a first approach, fluorescence labeling of AvBD7 allowed to track its systemic distribution after intraperitoneal injection in mice using whole body live imaging. It was associated to peritoneal cells and to deeper organs such as the liver. In the next step, the use of labeled AvBD7 allowed to observe its interaction with murine macrophages in culture. After incubation, it was able to penetrate inside the cells through an endocytosis-like mechanism. Furthermore, natural AvBD7 contributed to the control of intracellular multiplication of a multidrug resistant Salmonella strain, after incubation with infected macrophages. Finally, administration in a model of systemic lethal Salmonella infection in mice led to significant improvement of mouse survival, consistently with significant reduction of the liver bacterial load. In conclusion, the results reveal a hitherto unknown intracellular antibacterial effect of AvBD7 in Salmonella target cells and support AvBD7 as a candidate of interest for the treatment of infectious diseases caused by multidrug-resistant pathogenic Enterobacteriaceae.

19.
Artigo em Inglês | MEDLINE | ID: mdl-29276700

RESUMO

Salmonella is a facultative intracellular Gram-negative bacterium, responsible for a wide range of food- and water-borne diseases ranging from gastroenteritis to typhoid fever depending on hosts and serotypes. Salmonella thus represents a major threat to public health. A key step in Salmonella pathogenesis is the invasion of phagocytic and non-phagocytic host cells. To trigger its own internalization into non-phagocytic cells, Salmonella has developed different mechanisms, involving several invasion factors. For decades, it was accepted that Salmonella could only enter cells through a type three secretion system, called T3SS-1. Recent research has shown that this bacterium expresses outer membrane proteins, such as the Rck protein, which is able to induce Salmonella entry mechanism. Rck mimics natural host cell ligands and triggers engulfment of the bacterium by interacting with the epidermal growth factor receptor. Salmonella is thus able to use multiple entry pathways during the Salmonella infection process. However, it is unclear how and when Salmonella exploits the T3SS-1 and Rck entry mechanisms. As a series of reviews have focused on the T3SS-1, this review aims to describe the current knowledge and the limitations of our understanding of the Rck outer membrane protein. The regulatory cascade which controls Rck expression and the molecular mechanisms underlying Rck-mediated invasion into cells are summarized. The potential role of Rck-mediated invasion in Salmonella pathogenesis and the intracellular behavior of the bacteria following a Salmonella Rck-dependent entry are discussed.


Assuntos
Adesinas Bacterianas/metabolismo , Aderência Bacteriana , Endocitose , Infecções por Salmonella/microbiologia , Salmonella/fisiologia , Receptores ErbB/metabolismo , Regulação Bacteriana da Expressão Gênica , Ligação Proteica , Sistemas de Secreção Tipo III/metabolismo
20.
FEMS Microbiol Lett ; 361(1): 1-7, 2014 12.
Artigo em Inglês | MEDLINE | ID: mdl-25283734

RESUMO

Salmonella is a facultative intracellular bacterium found within a variety of phagocytic and nonphagocytic cells in vitro and in vivo For decades, it has been accepted that Salmonella can enter cells only through a Trigger mechanism mediated by a type three secretion system, called T3SS-1. However, recent researches have shown that this bacterium can use other invasion pathways mediating either Trigger or Zipper entry processes. Following eukaryotic cell invasion, Salmonella has to ensure its survival and proliferation within host cells. To do so, this bacterium resides either within a membrane-bound vacuole or freely within host cell cytosol. It is not clear why Salmonella has developed these alternate mechanisms for cell invasion and proliferation, but this provides a new insight into the mechanisms leading to Salmonella-induced diseases. Thus, the aim of this review is to show the evolution of Salmonella-host cell interaction paradigms by summarizing the different strategies used by Salmonella serotypes to invade and proliferate into eukaryotic cells.


Assuntos
Células Eucarióticas/microbiologia , Interações Hospedeiro-Patógeno , Salmonella/imunologia , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Modelos Biológicos , Sorogrupo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA