Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Med Virol ; 96(7): e29819, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39030992

RESUMO

Pregnant women represent a high-risk population for Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) infection. The presence of SARS-CoV-2 has been reported in placenta from infected pregnant women, but whether the virus influences placenta immune response remains unclear. We investigated the properties of maternal-fetal interface macrophages (MFMs) in a cohort of unvaccinated women who contracted coronavirus disease 2019 (COVID-19) during their pregnancy. We reported an infiltration of CD163+ macrophages in placenta from COVID-19 women 19 whereas lymphoid compartment was not affected. Isolated MFMs exhibited nonpolarized activated signature (NOS2, IDO1, IFNG, TNF, TGFB) mainly in women infected during the second trimester of pregnancy. COVID-19 during pregnancy primed MFM to produce type I and III interferon response to SARS-CoV-2 (Wuhan and δ strains), that were unable to elicit this in MFMs from healthy pregnant women. COVID-19 also primed SARS-CoV-2 internalization by MFM in an angiotensin-converting enzyme 2-dependent manner. Activation and recall responses of MFMs were influenced by fetal sex. Collectively, these findings support a role for MFMs in the local immune response to SARS-CoV-2 infection, provide a basis for protective placental immunity in COVID-19, and highlight the interest of vaccination in pregnant women.


Assuntos
COVID-19 , Macrófagos , Placenta , Complicações Infecciosas na Gravidez , SARS-CoV-2 , Humanos , Feminino , Gravidez , COVID-19/imunologia , COVID-19/virologia , Placenta/imunologia , Placenta/virologia , Macrófagos/imunologia , Macrófagos/virologia , Complicações Infecciosas na Gravidez/virologia , Complicações Infecciosas na Gravidez/imunologia , SARS-CoV-2/imunologia , Adulto , Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/metabolismo , Internalização do Vírus
2.
Cells ; 12(16)2023 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-37626837

RESUMO

Although very common, the precise mechanisms that explain the symptomatology of neuroendocrine syncope (NES) remain poorly understood. This disease, which can be very incapacitating, manifests itself as a drop in blood pressure secondary to vasodilation and/or extreme slowing of heart rate. As studies continue, the involvement of the adenosinergic system is becoming increasingly evident. Adenosine, which is an ATP derivative, may be involved in a large number of cases. Adenosine acts on G protein-coupled receptors with seven transmembrane domains. A1 and A2A adenosine receptor dysfunction seem to be particularly implicated since the activation leads to severe bradycardia or vasodilation, respectively, two cardinal symptoms of NES. This mini-review aims to shed light on the links between dysfunction of the adenosinergic system and NHS. In particular, signal transduction pathways through the modulation of cAMP production and ion channels in relation to effects on the cardiovascular system are addressed. A better understanding of these mechanisms could guide the pharmacological development of new therapeutic approaches.


Assuntos
Adenosina , Síncope , Animais , Anuros , Pressão Sanguínea , Frequência Cardíaca
3.
Int J Mol Sci ; 24(2)2023 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-36674725

RESUMO

Infection by Coxiella burnetii, the etiological agent of Q fever, poses the risk of causing severe obstetrical complications in pregnant women. C. burnetii is known for its placental tropism based on animal models of infection. The Nine Mile strain has been mostly used to study C. burnetii pathogenicity but the contribution of human isolates to C. burnetii pathogenicity is poorly understood. In this study, we compared five C. burnetii isolates from human placentas with C. burnetii strains including Nine Mile (NM) as reference. Comparative genomic analysis revealed that the Cb122 isolate was distinct from other placental isolates and the C. burnetii NM strain with a set of unique genes involved in energy generation and a type 1 secretion system. The infection of Balb/C mice with the Cb122 isolate showed higher virulence than that of NM or other placental isolates. We evaluated the pathogenicity of the Cb122 isolate by in vitro and ex vivo experiments. As C. burnetii is known to infect and survive within macrophages, we isolated monocytes and placental macrophages from healthy donors and infected them with the Cb122 isolate and the reference strain. We showed that bacteria from the Cb122 isolate were less internalized by monocyte-derived macrophages (MDM) than NM bacteria but the reference strain and the Cb122 isolate were similarly internalized by placental macrophages. The Cb122 isolate and the reference strain survived similarly in the two macrophage types. While the Cb122 isolate and the NM strain stimulated a poorly inflammatory program in MDM, they elicited an inflammatory program in placenta macrophages. We also reported that the Cb122 isolate and NM strain were internalized by trophoblastic cell lines and primary trophoblasts without specific replicative profiles. Placental explants were then infected with the Cb122 isolate and the NM strain. The bacteria from the Cb122 isolate were enriched in the chorionic villous foetal side. It is likely that the Cb122 isolate exhibited increased virulence in the multicellular environment provided by explants. Taken together, these results showed that the placental isolate of C. burnetii exhibits a specific infectious profile but its pathogenic role is not as high as the host immune response in pregnant women.


Assuntos
Coxiella burnetii , Febre Q , Animais , Camundongos , Feminino , Humanos , Gravidez , Coxiella burnetii/genética , Placenta/patologia , Macrófagos , Trofoblastos/patologia
4.
J Infect Dis ; 226(6): 1075-1083, 2022 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-35776143

RESUMO

BACKGROUND: Rickettsia felis is emergent in tropical areas. Despite its high morbidity, its natural history has not yet been fully determined. We investigated the role of the common household booklouse, Liposcelis bostrychophila, recently found to harbor R. felis. METHODS: Blood samples from 372 febrile patients from Senegalese villages, as well as nasal and skin samples from 264 asymptomatic individuals, were tested for cat flea-associated and booklice-associated strains of R. felis. Dust samples from beds were collected to isolate booklice and R. felis. Mice were infected with aerosol of R. felis strain from naturally infected booklice. RESULTS: Forty febrile patients (11%) were infected by R. felis, including 26 (7%) by the booklice-associated strain. Nine nasal samples (3.4%) and 28 skin samples (10.6%) contained R. felis, including 7 and 24, respectively, with the booklice-associated strain. The presence of live L. bostrychophila was observed in 32 dust samples (16.8%); R. felis was identified in 62 dust samples (32.5%). Several mice samples were positive for R. felis; interstitial lymphohistiocytic infiltrates were identified in lungs. CONCLUSIONS: Liposcelis bostrychophila may be a reservoir of R. felis. The booklice-associated strain is pathogenic in mammals, causing pneumonia. Human infection may be acquired via inhalation of infected booklice particles.


Assuntos
Felis , Pneumonia , Rickettsia felis , Animais , Poeira , Humanos , Mamíferos , Camundongos
5.
Front Microbiol ; 11: 1546, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32765448

RESUMO

T-bet is a transcription factor known to initiate and coordinate the gene expression program during Th1 differentiation, which is crucial for clearance of intracellular pathogens. Q fever is a worldwide zoonosis caused by Coxiella burnetii. This bacterium is transmitted to humans by aerosol. Indeed, the inhibition of the Coxiella-specific adaptive Th1 immune response leads to persistent infection and organ injury. How deficiency of T-bet affects host infection by C. burnetii has not been investigated. Here, using mice with a deletion of the T-bet gene and an airborne mode of infection to reproduce the natural conditions of C. burnetii infection, we show that infected T-bet-/- mice were more affected than wild-type mice. The lack of T-bet leads to defective bacterial control, intense replication, persistent infection, and organ injury manifesting as an increased number of granulomas. The absence of T-bet was also associated with an impaired immune response. Indeed, the production of the immunomodulatory cytokines interleukin (IL)-6 and IL-10 was increased, whereas the expression of microbicidal genes by splenocytes was impaired. Moreover, the absence of T-bet exhibited impaired production of interferon-γ, the principal cytokine released by Th1 effector cells. Thus, our study highlights the key role of T-bet in the control of C. burnetii infection in mice and leads to a reappraisal of granulomas in the pathogenesis of Q fever disease.

6.
Am J Trop Med Hyg ; 100(6): 1407-1412, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30938280

RESUMO

In recent years, bed bugs have reappeared in greater numbers, more frequently, and are biting humans in many new geographic areas. Infestations by these hematophagous insects are rapidly increasing worldwide. Borrelia recurrentis, a spirochete bacterium, is the etiologic agent of louse-borne relapsing fever. The known vectors are body lice, Pediculus humanus humanus. However, previous studies have suggested that bed bugs might also be able to transmit this bacterium. Adult Cimex lectularius were artificially infected with a blood meal mixed with bacterial suspension of B. recurrentis. They were subsequently fed with pathogen-free human blood until the end of the experiment. Bed bugs and feces were collected every 5 days to evaluate the capacity of bed bugs to acquire and excrete viable B. recurrentis using molecular biology, cultures, fluorescein diacetate and immunofluorescence assays. The feces collected on the day 5 and 10 postinfection contained viable bacteria. Immunofluorescence analysis of exposed bed bugs showed the presence of B. recurrentis in the digestive tract, even in bed bugs collected on day 20 after infection. Like human body lice, bed bugs can acquire, maintain, and excrete viable B. recurrentis that might infect humans through skin lesions. This preliminary work suggests that bed bugs might be competent vectors of B. recurrentis. Because bed bugs and body lice may share the same ecological niches, the role of bed bugs in transmitting recurrent fevers deserves further study.


Assuntos
Percevejos-de-Cama/microbiologia , Borrelia/fisiologia , Insetos Vetores/microbiologia , Febre Recorrente/transmissão , Animais , Sangue/microbiologia , Fezes/microbiologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C
7.
PLoS Genet ; 14(8): e1007569, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30080859

RESUMO

Ivermectin has emerged as very promising pediculicide, particularly in cases of resistance to commonly used pediculicides. Recently, however, the first field-evolved ivermectin-resistance in lice was reported. To gain insight into the mechanisms underlying ivermectin-resistance, we both looked for mutations in the ivermectin-target site (GluCl) and searched the entire proteome for potential new loci involved in resistance from laboratory susceptible and ivermectin-selected resistant body lice. Polymorphism analysis of cDNA GluCl showed no non-silent mutations. Proteomic analysis identified 22 differentially regulated proteins, of which 13 were upregulated and 9 were downregulated in the resistant strain. We evaluated the correlation between mRNA and protein levels by qRT-PCR and found that the trend in transcriptional variation was consistent with the proteomic changes. Among differentially expressed proteins, a complexin i.e. a neuronal protein which plays a key role in regulating neurotransmitter release, was shown to be the most significantly down-expressed in the ivermectin-resistant lice. Moreover, DNA-mutation analysis revealed that some complexin transcripts from resistant lice gained a premature stop codon, suggesting that this down-expression might be due, in part, to secondary effects of a nonsense mutation inside the gene. We further confirmed the association between complexin and ivermectin-resistance by RNA-interfering and found that knocking down the complexin expression induces resistance to ivermectin in susceptible lice. Our results provide evidence that complexin plays a significant role in regulating ivermectin resistance in body lice and represents the first evidence that links complexin to insecticide resistance.


Assuntos
Proteínas de Insetos/metabolismo , Resistência a Inseticidas/genética , Ivermectina , Proteínas do Tecido Nervoso/metabolismo , Pediculus/genética , Sequência de Aminoácidos , Animais , Clonagem Molecular , Regulação da Expressão Gênica , Proteínas de Insetos/genética , Inseticidas , Infestações por Piolhos/tratamento farmacológico , Proteínas do Tecido Nervoso/genética , Proteômica , RNA Mensageiro/genética , Análise de Sequência de DNA
8.
Clin Infect Dis ; 65(1): 1-5, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28379309

RESUMO

Background: Methanogens are antibiotic-resistant anaerobic archaea that escape routine detection in clinical microbiology. We hypothesized that methanogens are part of the anaerobic community that cause brain abscess. Methods: Methanogens were investigated in 1 index sample using specific polymerase chain reaction (PCR) sequencing and culture. The pathogenesis of a methanogen isolate was assessed in a mouse model. Archaea-specific quantitative (q) PCR and metagenomics were used to detect specific archaeal sequences in brain abscess samples and controls. Results: In 1 index sample, routine culture found Porphyromonas endodontalis and Streptococcus intermedius, and specific culture found Methanobrevibacter oralis susceptible to metronidazole and fusidic acid. Archaea-targeted PCR sequencing and metagenomics confirmed M. oralis along with 14 bacteria, including S. intermedius. Archaea-specific qPCR yielded archaea in 8/18 brain abscess specimens and 1/27 controls (P < .003), and metagenomics yielded archaea, mostly methanogens, in 28/32 brain abscess samples, and no archaea in 71 negative controls (P < 10-6). Infection of mice brains yielded no mortality in 14 controls and death in 17/22 M. oralis-inoculated mice (P < 10-6), 32/95 S. intermedius-inoculated mice (P < 10-6), and 75/104 mice inoculated with M. oralis mixed with S. intermedius (P < 10-6) 7 days post-inoculation. Conclusion: Methanogens belong to the anaerobic community responsible for brain abscess, and M. oralis may participate in the pathogenicity of this deadly infection. In mice, a synergy of M. oralis and S. intermedius was observed. Antibiotic treatment of brain abscess should contain anti-archaeal compounds such as imidazole derivatives in most cases.


Assuntos
Abscesso Encefálico/microbiologia , Methanobrevibacter/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Abscesso Encefálico/mortalidade , Criança , Pré-Escolar , DNA Arqueal/genética , DNA Bacteriano/genética , Modelos Animais de Doenças , Feminino , Humanos , Lactente , Recém-Nascido , Camundongos , Pessoa de Meia-Idade , Porphyromonas endodontalis/genética , Reação em Cadeia da Polimerase em Tempo Real , Estudos Retrospectivos , Streptococcus intermedius/genética , Adulto Jovem
9.
Artigo em Inglês | MEDLINE | ID: mdl-28131375

RESUMO

Aerosols from the products of abortions of infected animals with Coxiella burnetii were known to be the main source of infection in humans with this bacterium. However, little is known about the excretion of C. burnetii in the feces and urine of infected mice, or the dynamic of transmission between infected and healthy mice. To investigate whether C. burnetii can be excreted in the feces and urine of infected mice and whether transmission to uninfected cage mates occurs, male mice were inoculated with C. burnetii using a "whole body aerosol system" and feces and urine were collected different time points post-infection from these mice. One hour post exposure to aerosols, uninfected mice was placed with infected mice and the transmission was monitored using blood, and organs biopsies collected after sacrifice of contact mice different time points post-contact. Bacterial DNA was not detected in the feces and urine of infected mice at 3, 7, 14 and 28days post-inoculation suggesting that C. burnetii was not excreted in the feces and urine and consequently they cannot be source of contamination. However, based on the positive PCR results for lungs, blood, spleen, tracheal lymph nodes and cervical lymph nodes, some of the contact animals were considered contaminated at 8days post-contact. These results indicated that transmission of C. burnetii to contact animals occurs, and it is unlikely that feces and urine act as source of this transmission. Further experiments are needed to clarify the exact mode of contamination.


Assuntos
Coxiella burnetii/isolamento & purificação , Fezes/microbiologia , Febre Q/transmissão , Aerossóis/administração & dosagem , Animais , Anticorpos Antibacterianos/sangue , Coxiella burnetii/genética , Coxiella burnetii/patogenicidade , DNA Bacteriano/análise , Modelos Animais de Doenças , Masculino , Camundongos , Reação em Cadeia da Polimerase , Febre Q/microbiologia , Urina/microbiologia
10.
Malar J ; 16(1): 5, 2017 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-28049524

RESUMO

BACKGROUND: Malaria is still a major public health issue worldwide, and one of the best approaches to fight the disease remains vector control. The current methods for mosquito identification include morphological methods that are generally time-consuming and require expertise, and molecular methods that require laboratory facilities with relatively expensive running costs. Matrix-Assisted Laser Desorption Ionization Time-Of-Flight Mass Spectrometry (MALDI-TOF MS) technology, routinely used for bacterial identification, has recently emerged in the field of entomology. The aim of the present study was to assess whether MALDI-TOF MS could successfully distinguish Anopheles stephensi mosquitoes according to their Plasmodium infection status. METHODS: C57BL/6 mice experimentally infected with Plasmodium berghei were exposed to An. stephensi bites. For the determination of An. stephensi infection status, mosquito cephalothoraxes were dissected and submitted to mass spectrometry analyses and DNA amplification for molecular analysis. Spectra were grouped according to mosquitoes' infection status and spectra quality was validated based on intensity and reproducibility within each group. The in-lab MALDI-TOF MS arthropod reference spectra database, upgraded with representative spectra from both groups (infected/non-infected), was subsequently queried blindly with cephalothorax spectra from specimens of both groups. RESULTS: The MALDI TOF MS profiles generated from protein extracts prepared from the cephalothorax of An. stephensi allowed distinction between infected and uninfected mosquitoes. Correct classification was obtained in blind test analysis for (79/80) 98.75% of all mosquitoes tested. Only one of 80 specimens, an infected mosquito, was misclassified in the blind test analysis. CONCLUSIONS: Matrix-Assisted Laser Desorption Ionization Time-Of-Flight Mass Spectrometry appears to be a promising, rapid and reliable tool for the epidemiological surveillance of Anopheles vectors, including their identification and their infection status.


Assuntos
Anopheles/parasitologia , Produtos Biológicos/análise , Entomologia/métodos , Plasmodium berghei/química , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Animais , Anopheles/crescimento & desenvolvimento , Feminino , Camundongos Endogâmicos C57BL
11.
Microbiol Spectr ; 4(4)2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27726780

RESUMO

Epidemic typhus caused by Rickettsia prowazekii is one of the oldest pestilential diseases of humankind. The disease is transmitted to human beings by the body louse Pediculus humanus corporis and is still considered a major threat by public health authorities, despite the efficacy of antibiotics, because poor sanitary conditions are conducive to louse proliferation. Epidemic typhus has accompanied disasters that impact humanity and has arguably determined the outcome of more wars than have soldiers and generals. The detection, identification, and characterization of microorganisms in ancient remains by paleomicrobiology has permitted the diagnosis of past epidemic typhus outbreaks through the detection of R. prowazekii. Various techniques, including microscopy and immunodetection, can be used in paleomicrobiology, but most of the data have been obtained by using PCR-based molecular techniques on dental pulp samples. Paleomicrobiology enabled the identification of the first outbreak of epidemic typhus in the 18th century in the context of a pan-European great war in the city of Douai, France, and supported the hypothesis that typhus was imported into Europe by Spanish soldiers returning from America. R. prowazekii was also detected in the remains of soldiers of Napoleon's Grand Army in Vilnius, Lithuania, which indicates that Napoleon's soldiers had epidemic typhus. The purpose of this article is to underscore the modern comprehension of clinical epidemic typhus, focus on the historical relationships of the disease, and examine the use of paleomicrobiology in the detection of past epidemic typhus outbreaks.


Assuntos
Surtos de Doenças , Fósseis/microbiologia , Paleopatologia/métodos , Rickettsia prowazekii/isolamento & purificação , Tifo Epidêmico Transmitido por Piolhos/epidemiologia , Tifo Epidêmico Transmitido por Piolhos/história , Animais , Técnicas Bacteriológicas/métodos , Saúde Global , História do Século XVIII , História do Século XIX , História do Século XX , História do Século XXI , Humanos , Pediculus/crescimento & desenvolvimento , Pediculus/microbiologia , Reação em Cadeia da Polimerase/métodos , Rickettsia prowazekii/genética
12.
Infect Immun ; 84(7): 2116-2123, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27160294

RESUMO

Coxiella burnetii is mainly transmitted by aerosols and is responsible for multiple-organ lesions. Animal models have shown C. burnetii pathogenicity, but long-term outcomes still need to be clarified. We used a whole-body aerosol inhalation exposure system to mimic the natural route of infection in immunocompetent (BALB/c) and severe combined immunodeficient (SCID) mice. After an initial lung inoculum of 10(4) C. burnetii cells/lung, the outcome, serological response, hematological disorders, and deep organ lesions were described up to 3 months postinfection. C. burnetii-specific PCR, anti-C. burnetii immunohistochemistry, and fluorescent in situ hybridization (FISH) targeting C. burnetii-specific 16S rRNA completed the detection of the bacterium in the tissues. In BALB/c mice, a thrombocytopenia and lymphopenia were first observed, prior to evidence of C. burnetii replication. In all SCID mouse organs, DNA copies increased to higher levels over time than in BALB/c ones. Clinical signs of discomfort appeared in SCID mice, so follow-up had to be shortened to 2 months in this group. At this stage, all animals presented bone, cervical, and heart lesions. The presence of C. burnetii could be attested in situ for all organs sampled using immunohistochemistry and FISH. This mouse model described C. burnetii Nine Mile strain spread using aerosolization in a way that corroborates the pathogenicity of Q fever described in humans and completes previously published data in mouse models. C. burnetii infection occurring after aerosolization in mice thus seems to be a useful tool to compare the pathogenicity of different strains of C. burnetii.


Assuntos
Coxiella burnetii , Febre Q/microbiologia , Febre Q/transmissão , Aerossóis , Animais , Contagem de Células Sanguíneas , Coxiella burnetii/genética , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Fenótipo , Febre Q/diagnóstico
13.
Proc Natl Acad Sci U S A ; 112(26): 8088-93, 2015 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-26056256

RESUMO

A growing number of recent reports have implicated Rickettsia felis as a human pathogen, paralleling the increasing detection of R. felis in arthropod hosts across the globe, primarily in fleas. Here Anopheles gambiae mosquitoes, the primary malarial vectors in sub-Saharan Africa, were fed with either blood meal infected with R. felis or infected cellular media administered in membrane feeding systems. In addition, a group of mosquitoes was fed on R. felis-infected BALB/c mice. The acquisition and persistence of R. felis in mosquitoes was demonstrated by quantitative PCR detection of the bacteria up to day 15 postinfection. R. felis was detected in mosquito feces up to day 14. Furthermore, R. felis was visualized by immunofluorescence in salivary glands, in and around the gut, and in the ovaries, although no vertical transmission was observed. R. felis was also found in the cotton used for sucrose feeding after the mosquitoes were fed infected blood. Natural bites from R. felis-infected An. gambiae were able to cause transient rickettsemias in mice, indicating that this mosquito species has the potential to be a vector of R. felis infection. This is particularly important given the recent report of high prevalence of R. felis infection in patients with "fever of unknown origin" in malaria-endemic areas.


Assuntos
Anopheles/microbiologia , Insetos Vetores , Infecções por Rickettsia/transmissão , Rickettsia felis/patogenicidade , Animais , Modelos Animais de Doenças , Fezes/microbiologia , Feminino , Imunofluorescência , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Rickettsia/microbiologia
14.
Cell Mol Life Sci ; 72(14): 2739-48, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25725801

RESUMO

The CD28 costimulatory receptor has a pivotal role in T cell biology as this molecule amplifies T cell receptor (TCR) signals to provide an efficient immune T cell response. There is a large debate about how CD28 mediates these signals. Here, we designed a CD28 gene-targeted knock-in mouse strain lacking the cytoplasmic tail of CD28. As is the case in CD28-deficient (CD28 knock-out) mice, regulatory T cell homeostasis and T cell activation are altered in these CD28 knock-in mice. Unexpectedly, the presence of a CD28 molecule deprived of its cytoplasmic tail could partially induce some early activation events in T cells such as signaling events or expression of early activation markers. These results unravel a new mechanism of T cell costimulation by CD28, independent of its cytoplasmic tail.


Assuntos
Antígenos CD28/fisiologia , Ativação Linfocitária/fisiologia , Linfócitos T/imunologia , Animais , Antígenos CD28/química , Antígenos CD28/genética , Técnicas de Introdução de Genes , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Estrutura Terciária de Proteína , Febre Q/imunologia , Transdução de Sinais
15.
PLoS Pathog ; 11(3): e1004732, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25781937

RESUMO

The detection of the activities of pathogen-encoded virulence factors by the innate immune system has emerged as a new paradigm of pathogen recognition. Much remains to be determined with regard to the molecular and cellular components contributing to this defense mechanism in mammals and importance during infection. Here, we reveal the central role of the IL-1ß signaling axis and Gr1+ cells in controlling the Escherichia coli burden in the blood in response to the sensing of the Rho GTPase-activating toxin CNF1. Consistently, this innate immune response is abrogated in caspase-1/11-impaired mice or following the treatment of infected mice with an IL-1ß antagonist. In vitro experiments further revealed the synergistic effects of CNF1 and LPS in promoting the maturation/secretion of IL-1ß and establishing the roles of Rac, ASC and caspase-1 in this pathway. Furthermore, we found that the α-hemolysin toxin inhibits IL-1ß secretion without affecting the recruitment of Gr1+ cells. Here, we report the first example of anti-virulence-triggered immunity counteracted by a pore-forming toxin during bacteremia.


Assuntos
Toxinas Bacterianas/imunologia , Infecções por Escherichia coli/imunologia , Proteínas de Escherichia coli/imunologia , Proteínas Hemolisinas/imunologia , Imunidade Inata/imunologia , Transdução de Sinais/imunologia , Animais , Bacteriemia/imunologia , Modelos Animais de Doenças , Escherichia coli/imunologia , Escherichia coli/patogenicidade , Feminino , Interações Hospedeiro-Patógeno/imunologia , Interleucina-1beta/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Virulência , Fatores de Virulência/imunologia
16.
Microb Pathog ; 80: 1-6, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25680834

RESUMO

Programmed death ligand-1 (PD-L1) is a co-signaling molecule that regulates T-cell responses in vivo. Its role in bacterial infections, including Q fever, a zoonosis due to Coxiella burnetii infection, is not well understood. We showed by flow cytometry that PD-L1 membrane expression was specifically increased in T-cells from patients with acute Q fever, not from patients with Q fever endocarditis, suggesting that PD-L1 plays a role in the early phases of C. burnetii infection. To assess this hypothesis, we studied the role of PD-L1 in C. burnetii-infected mice. C. burnetii infection resulted in PD-L1 up-regulation in splenocytes. Anti-PD-L1 antibodies injected into the mice did not affect the total number of splenic T-cells but increased the relative number of CD4(+) T-cells compared with CD8(+) T-cells. Additionally, anti-PD-L1 antibodies significantly increased the number of splenic CD4(+) and CD8(+) T cells that expressed low membrane CD62L levels. Our results indicate that the increased expression of PD-L1 by T-cells is associated with a decreased number of memory T-cells during C. burnetii infection, opening new perspectives in the understanding of Q fever pathophysiology.


Assuntos
Antígeno B7-H1/análise , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Memória Imunológica , Febre Q/imunologia , Subpopulações de Linfócitos T/imunologia , Regulação para Cima , Animais , Feminino , Citometria de Fluxo , Imunofenotipagem , Selectina L/análise , Contagem de Linfócitos , Camundongos Endogâmicos C57BL
17.
PLoS One ; 9(5): e97503, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24835240

RESUMO

Coxiella burnetii, the agent of Q fever, is known to persist in humans and rodents but its cellular reservoir in hosts remains undetermined. We hypothesized that adipose tissue serves as a C. burnetii reservoir during bacterial latency. BALB/c and C57BL/6 mice were infected with C. burnetii by the intraperitoneal route or the intracheal route. Adipose tissue was tested for the presence of C. burnetii several months after infection. C. burnetii was detected in abdominal, inguinal and dorsal adipose tissue 4 months post-infection, when no bacteria were detected in blood, liver, lungs and spleen, regardless of the inoculation route and independently of mouse strain. The transfer of abdominal adipose tissue from convalescent BALB/c mice to naïve immunodeficient mice resulted in the infection of the recipient animals. It is likely that C. burnetii infects adipocytes in vivo because bacteria were found in adipocytes within adipose tissue and replicated within in vitro-differentiated adipocytes. In addition, C. burnetii induced a specific transcriptional program in in-vivo and in vitro-differentiated adipocytes, which was enriched in categories associated with inflammatory response, hormone response and cytoskeleton. These changes may account for bacterial replication in in-vitro and chronic infection in-vivo. Adipose tissue may be the reservoir in which C. burnetii persists for prolonged periods after apparent clinical cure. The mouse model of C. burnetii infection may be used to understand the relapses of Q fever and provide new perspectives to the follow-up of patients.


Assuntos
Tecido Adiposo/microbiologia , Coxiella burnetii , Reservatórios de Doenças , Febre Q/microbiologia , Tecido Adiposo/fisiologia , Animais , Diferenciação Celular/fisiologia , Primers do DNA/genética , Feminino , Imunofluorescência , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Análise em Microsséries , Reação em Cadeia da Polimerase , RNA Ribossômico 16S/genética , Reação em Cadeia da Polimerase em Tempo Real
18.
Artigo em Inglês | MEDLINE | ID: mdl-25688336

RESUMO

Pediculus humanus humanus is an human ectoparasite which represents a serious public health threat because it is vector for pathogenic bacteria. It is important to understand and identify where bacteria reside in human body lice to define new strategies to counterstroke the capacity of vectorization of the bacterial pathogens by body lice. It is known that phagocytes from vertebrates can be hosts or reservoirs for several microbes. Therefore, we wondered if Pediculus humanus humanus phagocytes could hide pathogens. In this study, we characterized the phagocytes from Pediculus humanus humanus and evaluated their contribution as hosts for human pathogens such as Rickettsia prowazekii, Bartonella Quintana, and Acinetobacter baumannii.


Assuntos
Acinetobacter baumannii/fisiologia , Infecções Bacterianas/microbiologia , Bartonella quintana/fisiologia , Insetos Vetores/microbiologia , Pediculus/microbiologia , Rickettsia prowazekii/fisiologia , Animais , Infecções Bacterianas/transmissão , Reservatórios de Doenças/microbiologia , Hemócitos/microbiologia , Humanos , Fagócitos/microbiologia
19.
Comp Immunol Microbiol Infect Dis ; 35(6): 551-6, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22784931

RESUMO

Pathogenic rickettsiae of the spotted fever group (SFG) induce skin lesions called "eschars" at the sites of arthropod bites. We recently described a guinea pig model based on eschar formation to predict the eventual pathogenicity of new Rickettsia species, and in this study, we used the model to study cross-protection among SFG Rickettsia species and subspecies. We showed that the intradermal inoculation of guinea pigs with Rickettsia conorii subsp. conorii significantly reduced, but not completely prevented, the number of eschars after sub-challenges with the same pathogen (P=0.0004). The same effect was also observed for other subspecies of the R. conorii complex. Additionally, the bacterial load in the eschars was significantly lower in immunized animals than in naïve animals. No protection was observed when sub-challenges were performed with other Rickettsia species, such as R. africae, R. sibirica subsp. mongolitimonae, R. aeschlimannii and R. massiliae. Our data suggest that patients may experience several episodes of infection with related or with the same species of Rickettsia. Moreover, the absence of cross-protection between Rickettsia species may explain the co-existence of two or more tick-borne rickettsioses in the same geographic areas.


Assuntos
Febre Botonosa/imunologia , Rickettsia/imunologia , Dermatopatias Bacterianas/imunologia , Animais , Febre Botonosa/microbiologia , Proteção Cruzada , DNA Bacteriano/genética , Modelos Animais de Doenças , Feminino , Cobaias , Humanos , Rickettsia/classificação , Rickettsia/genética , Rickettsia/patogenicidade , Rickettsia conorii/genética , Rickettsia conorii/imunologia , Rickettsia conorii/patogenicidade , Dermatopatias Bacterianas/microbiologia
20.
Adv Exp Med Biol ; 984: 287-98, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22711638

RESUMO

Coxiella burnetii, the causative agent of Q fever, has evolved a wealth of mechanisms in order to persist within hosts. Two tissues, namely adipose tissue and placenta, are candidates to house C. burnetii, but the mechanisms governing C. burnetii survival in these tissues are still unknown. In contrast, monocytes and macrophages are well-known targets of C. burnetii. First, C. burnetii has developed a specific strategy of phagocytosis subversion that consists of the inhibition of integrin interplay. Second, C. burnetii persistence is associated with macrophage activation profiles. Indeed, monocytes (in which C. burnetii survives without replication) exhibit a proinflammatory M1-type response, whereas macrophages (in which C. burnetii slowly replicates) are polarized towards an M2-type. Third, interleukin-10 produced by monocytes is a main factor of the chronic development of Q fever, and murine models confirm the key role of interleukin-10 in C. burnetii persistence. Fourth, apoptotic cells may play a key role in chronic Q fever. The uptake of apoptotic cells by circulating monocytes increases C. burnetii replication by redirecting monocytes toward a non-protective M2 profile. In the presence of interferon-γ, apoptotic cell engulfment is inhibited and monocytes polarized toward an M1 program are able to kill C. burnetii; this is the situation observed in patients with uncomplicated acute Q fever. Finally, we cannot exclude that regulatory T cells may play a role in C. burnetii persistence because their number is increased in patients with chronic Q fever.


Assuntos
Coxiella burnetii/imunologia , Febre Q/imunologia , Animais , Humanos , Interferon gama/imunologia , Interleucina-10/imunologia , Febre Q/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...