Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Infect Immun ; 91(2): e0032322, 2023 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-36688662

RESUMO

Dendritic cells (DCs) belong to the first line of innate defense and come into early contact with invading pathogens, including the zoonotic bacterium Coxiella burnetii, the causative agent of Q fever. However, the pathogen-host cell interactions in C. burnetii-infected DCs, particularly the role of mechanisms of immune subversion beyond virulent phase I lipopolysaccharide (LPS), as well as the contribution of cellular self-defense strategies, are not understood. Using phase II Coxiella-infected DCs, we show that impairment of DC maturation and MHC I downregulation is caused by autocrine release and action of immunosuppressive transforming growth factor-ß (TGF-ß). Our study demonstrates that IFN-γ reverses TGF-ß impairment of maturation/MHC I presentation in infected DCs and activates bacterial elimination, predominantly by inducing iNOS/NO. Induced NO synthesis strongly affects bacterial growth and infectivity. Moreover, our studies hint that Coxiella-infected DCs might be able to protect themselves from mitotoxic NO by switching from oxidative phosphorylation to glycolysis, thus ensuring survival in self-defense against C. burnetii. Our results provide new insights into DC subversion by Coxiella and the IFN-γ-mediated targeting of C. burnetii during early steps in the innate immune response.


Assuntos
Coxiella burnetii , Febre Q , Humanos , Fator de Crescimento Transformador beta , Febre Q/microbiologia , Interferon gama , Células Dendríticas
2.
Int J Med Microbiol ; 313(6): 151590, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38056089

RESUMO

The Q-GAPS (Q fever GermAn interdisciplinary Program for reSearch) consortium was launched in 2017 as a German consortium of more than 20 scientists with exceptional expertise, competence, and substantial knowledge in the field of the Q fever pathogen Coxiella (C.) burnetii. C. burnetii exemplifies as a zoonotic pathogen the challenges of zoonotic disease control and prophylaxis in human, animal, and environmental settings in a One Health approach. An interdisciplinary approach to studying the pathogen is essential to address unresolved questions about the epidemiology, immunology, pathogenesis, surveillance, and control of C. burnetii. In more than five years, Q-GAPS has provided new insights into pathogenicity and interaction with host defense mechanisms. The consortium has also investigated vaccine efficacy and application in animal reservoirs and identified expanded phenotypic and genotypic characteristics of C. burnetii and their epidemiological significance. In addition, conceptual principles for controlling, surveilling, and preventing zoonotic Q fever infections were developed and prepared for specific target groups. All findings have been continuously integrated into a Web-based, interactive, freely accessible knowledge and information platform (www.q-gaps.de), which also contains Q fever guidelines to support public health institutions in controlling and preventing Q fever. In this review, we will summarize our results and show an example of how an interdisciplinary consortium provides knowledge and better tools to control a zoonotic pathogen at the national level.


Assuntos
Coxiella burnetii , Saúde Única , Febre Q , Animais , Humanos , Coxiella burnetii/genética , Febre Q/epidemiologia , Febre Q/prevenção & controle , Zoonoses/epidemiologia , Zoonoses/prevenção & controle , Estudos Interdisciplinares
3.
Infect Immun ; 88(11)2020 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-32817330

RESUMO

Natural killer (NK) cells are critically involved in the early immune response against various intracellular pathogens, including Coxiella burnetii and Chlamydia psittaciChlamydia-infected NK cells functionally mature, induce cellular immunity, and protect themselves by killing the bacteria in secreted granules. Here, we report that infected NK cells do not allow intracellular multiday growth of Coxiella, as is usually observed in other host cell types. C. burnetii-infected NK cells display maturation and gamma interferon (IFN-γ) secretion, as well as the release of Coxiella-containing lytic granules. Thus, NK cells possess a potent program to restrain and expel different types of invading bacteria via degranulation. Strikingly, though, in contrast to Chlamydia, expulsed Coxiella organisms largely retain their infectivity and, hence, escape the cell-autonomous self-defense mechanism in NK cells.


Assuntos
Degranulação Celular/imunologia , Imunidade Celular/imunologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/microbiologia , Febre Q/imunologia , Animais , Coxiella burnetii , Camundongos
4.
Infect Immun ; 88(1)2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31658957

RESUMO

Dendritic cells (DCs) and natural killer (NK) cells are critically involved in the early response against various bacterial microbes. Functional activation of infected DCs and NK cell-mediated gamma interferon (IFN-γ) secretion essentially contribute to the protective immunity against Chlamydia How DCs and NK cells cooperate during the antichlamydial response is not fully understood. Therefore, in the present study, we investigated the functional interplay between Chlamydia-infected DCs and NK cells. Our biochemical and cell biological experiments show that Chlamydia psittaci-infected DCs display enhanced exosome release. We find that such extracellular vesicles (referred to as dexosomes) do not contain infectious bacterial material but strongly induce IFN-γ production by NK cells. This directly affects C. psittaci growth in infected target cells. Furthermore, NK cell-released IFN-γ in cooperation with tumor necrosis factor alpha (TNF-α) and/or dexosomes augments apoptosis of both noninfected and infected epithelial cells. Thus, the combined effect of dexosomes and proinflammatory cytokines restricts C. psittaci growth and attenuates bacterial subversion of apoptotic host cell death. In conclusion, this provides new insights into the functional cooperation between DCs, dexosomes, and NK cells in the early steps of antichlamydial defense.


Assuntos
Comunicação Celular , Infecções por Chlamydia/imunologia , Chlamydophila psittaci/imunologia , Células Dendríticas/metabolismo , Exossomos/metabolismo , Imunidade Inata , Células Matadoras Naturais/metabolismo , Animais , Células Cultivadas , Fatores Imunológicos/metabolismo , Interferon gama/metabolismo , Camundongos , Modelos Teóricos
5.
Vaccine ; 41(33): 4798-4807, 2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37357077

RESUMO

An inactivated Coxiella burnetii Phase I (PhI) vaccine (Coxevac®) is licensed in several European countries for goats and cattle to prevent coxiellosis. The vaccine is also applied to sheep, although detailed information about the ovine immune response and vaccine dose is missing. Eighteen gimmers from a C. burnetii unsuspected flock were randomly divided into three groups of six. Group 1 (Cox1) and 2 (Cox2) were vaccinated twice with 1 ml and 2 ml Coxevac®, respectively, three weeks apart (primary vaccination). The same procedure was applied with Cox3 (2 ml sodium chloride, control group). A third injection (booster) was performed after nine months. Potential side effects were determined by measuring the rectal body temperature and skin thickness at the injection site. Blood samples were collected to detect phase-specific IgM and IgG antibodies and interferon-É£ (IFN-É£) release by immunofluorescence assay and ELISAs, respectively. Moreover, a cell infection neutralization assay determined the appearance of neutralizing sera. Body temperatures increased for one day post vaccination, and the skin swelled only slightly. Regardless of the vaccine volume, immunized sheep reacted first with an IgM and IgG PhII response. Ten weeks after the primary vaccination, IgG PhI antibodies predominated. Boosting eight months after primary vaccination resulted in a robust IgG PhI increase and strong IFN-É£ response. In the vaccinated animals, the neutralizing effect is more widespread after the administration of 1 ml than after the treatment with 2 ml. In summary, differences between 1 and 2 ml Coxevac® are minor, and a vaccine volume of 1 ml seems to be sufficient. A booster after the primary vaccination is apparently necessary to stimulate the cell-mediated immune response in naïve sheep.


Assuntos
Coxiella burnetii , Febre Q , Animais , Ovinos , Bovinos , Febre Q/prevenção & controle , Febre Q/veterinária , Vacinas de Produtos Inativados , Vacinas Bacterianas , Imunidade Celular , Vacinação/veterinária , Vacinação/métodos , Interferon gama , Cabras , Imunoglobulina G , Imunoglobulina M
6.
Microbiol Spectr ; 10(6): e0281722, 2022 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-36219107

RESUMO

Tunneling nanotubes (TNTs) are transient cellular connections that consist of dynamic membrane protrusions. They play an important role in cell-to-cell communication and mediate the intercellular exchanges of molecules and organelles. TNTs can form between different cell types and may contribute to the spread of pathogens by serving as cytoplasmic corridors. We demonstrate that Chlamydia (C.) trachomatis-infected human embryonic kidney (HEK) 293 cells and other cells form TNT-like structures through which reticulate bodies (RBs) pass into uninfected cells. Observed TNTs have a life span of 1 to 5 h and contain microtubules, which are essential for chlamydial transfer. They can bridge distances of up to 50 µm between connecting neighboring cells. Consistent with the biological role for TNTs, we show that C. trachomatis spread also occurs under conditions in which the extracellular route of chlamydial entry into host cells is blocked. Based on our findings, we propose that TNTs play a critical role in the direct, cell-to-cell transmission of chlamydia. IMPORTANCE Intracellular bacterial pathogens often undergo a life cycle in which they parasitize infected host cells in membranous vacuoles. Two pathways have been described by which chlamydia can exit infected host cells: lytic cell destruction or exit via extrusion formation. Whether direct, cell-to-cell contact may also play a role in the spread of infection is unknown. Tunneling nanotubes (TNTs) interconnect the cytoplasm of adjacent cells to mediate efficient communication and the exchange of material between them. We used Chlamydia trachomatis and immortalized cells to analyze whether TNTs mediate bacterial transmission from an infected donor to uninfected acceptor cells. We show that chlamydia-infected cells build TNTs through which the intracellular reticulate bodies (RBs) of the chlamydia can pass into uninfected neighboring cells. Our study contributes to the understanding of the function of TNTs in the cell-to-cell transmission of intracellular pathogens and provides new insights into the strategies by which chlamydia spreads among multicellular tissues.


Assuntos
Chlamydia trachomatis , Nanotubos , Humanos , Células HEK293 , Comunicação Celular , Nanotubos/química
7.
Methods Protoc ; 4(2)2021 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-33917811

RESUMO

We present a new and straightforward method by which standard cell culture plates can be sealed off from ambient air and be placed under controlled hypoxic cell culture conditions without costly or highly specialized materials. The method was established on a murine cell culture system using the dendritic cell line JAWS II but can be readily adapted to other cell cultures. The procedure was designed to be easy to implement in cell culture laboratories with standard incubators and requires only readily available materials, resources, and consumables, such as six-well plates, degassed culture medium, CoCl2, a vacuum sealer, etc., and no further complicated laboratory equipment. The simple hypoxic cell culture method presented here is technically reliable and experimentally safe. As it can be performed in any standard incubator, it is suitable for use at both low and higher biosafety levels.

8.
Vet Sci ; 8(11)2021 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-34822625

RESUMO

A Q fever outbreak on a dairy goat and cattle farm was investigated with regard to the One Health concept. Serum samples and vaginal swabs from goats with different reproductive statuses were collected. Cows, cats, and a dog were investigated with the same sample matrix. The farmer's family was examined by serum samples. Ruminant sera were analyzed with two phase-specific enzyme-linked immunoassays (ELISAs). Dominant immunoglobulin G (IgG) phase II levels reflected current infections in goats. The cows had high IgG phase I and II levels indicating ongoing infections. Feline, canine, and human sera tested positive by indirect fluorescent antibody test (IFAT). Animal vaginal swabs were analyzed by qPCR to detect C. burnetii, and almost all tested positive. A new cattle-associated C. burnetii genotype C16 was identified by the Multiple-Locus Variable-number tandem repeat Analysis (MLVA/VNTR) from ruminant samples. Additionally, a possible influence of 17ß-estradiol on C. burnetii antibody response was evaluated in goat sera. Goats in early/mid-pregnancy had significantly lower levels of phase-specific IgGs and 17ß-estradiol than goats in late pregnancy. We conclude that the cattle herd may have transmitted C. burnetii to the pregnant goat herd, resulting in a Q fever outbreak with one acute human case. The influence of placentation and maternal pregnancy hormones during pregnancy on the immune response is discussed.

9.
Vaccine ; 39(10): 1499-1507, 2021 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-33558108

RESUMO

Qfever is a zoonotic disease caused by the bacterium Coxiella burnetii; Coxiella-infected ruminants are the main reservoir shedding the pathogen during abortion or parturition through birth products. Germany has a long history of small-scale Q fever epidemics in the human population mostly associated with lambing sheep. Therefore, fast and efficient control measures are essentially required to prevent transmission from infected sheep flocks to humans. In our present study, three sheep flocks were vaccinated with an inactivated C.burnetii phase I vaccine after a field infection with C.burnetii was diagnosed. Serum samples and vaginal swabs were collected at different time points to evaluate the extent of the outbreak and the consequences of the vaccination. The serum samples were examined by phase-specific IgG phase I and phase II ELISAs and a commercial ELISA, simultaneously detecting both phase variations. Moreover, vaginal swabs were analysed by qPCR. The fourth flock with no Q fever history and non-vaccinated animals were used as a control group to evaluate the phase-specific ELISAs. The inactivated C.burnetii phase I vaccine induced an IgG phase II response and boosted the humoral immune reaction against natural pre-infections. Furthermore, the longevity of vaccine-induced antibodies seems to depend on previous infections. Around 16 months after primary vaccination, mainly IgG phase I antibodies were detectable. Vaccination did not prevent shedding at the next lambing season. Most interestingly, the phase-specific ELISAs revealed more C.burnetii positive animals than the blended ELISA-Assay. Taken together, phase-specific ELISAs are suitable tools to provide insights into natural- or vaccine-induced humoral immune responses to C.burnetii in sheep.


Assuntos
Coxiella burnetii , Febre Q , Doenças dos Ovinos , Animais , Feminino , Alemanha , Cabras , Imunidade Humoral , Gravidez , Febre Q/prevenção & controle , Febre Q/veterinária , Ovinos , Doenças dos Ovinos/prevenção & controle , Vacinação/veterinária
10.
Sci Rep ; 9(1): 4799, 2019 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-30886314

RESUMO

Natural killer (NK) cells are innate immune cells critically involved in the early immune response against various pathogens including chlamydia. Here, we demonstrate that chlamydia-infected NK cells prevent the intracellular establishment and growth of the bacteria. Upon infection, they display functional maturation characterized by enhanced IFN-γ secretion, CD146 induction, PKCϴ activation, and granule secretion. Eventually, chlamydia are released in a non-infectious, highly immunogenic form driving a potent Th1 immune response. Further, anti-chlamydial antibodies generated during immunization neutralize the infection of epithelial cells. The release of chlamydia from NK cells requires PKCϴ function and active degranulation, while granule-associated granzyme B drives the loss of chlamydial infectivity. Cellular infection and bacterial release can be undergone repeatedly and do not affect NK cell function. Strikingly, NK cells passing through such an infection cycle significantly improve their cytotoxicity. Thus, NK cells not only protect themselves against productive chlamydial infections but also actively trigger potent anti-bacterial responses.


Assuntos
Chlamydophila psittaci/imunologia , Imunidade Celular , Células Matadoras Naturais/imunologia , Psitacose/imunologia , Células Th1/imunologia , Animais , Antígeno CD146/metabolismo , Comunicação Celular/imunologia , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Interferon gama/metabolismo , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/microbiologia , Camundongos , Cultura Primária de Células , Proteína Quinase C-theta/metabolismo , Psitacose/sangue , Psitacose/microbiologia , Baço/citologia
11.
Cell Rep ; 28(1): 30-38.e5, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31269448

RESUMO

Malaria represents a major cause of death from infectious disease. Hemozoin is a Plasmodium-derived product that contributes to progression of cerebral malaria. However, there is a gap of knowledge regarding how hemozoin is recognized by innate immunity. Myeloid C-type lectin receptors (CLRs) encompass a family of carbohydrate-binding receptors that act as pattern recognition receptors in innate immunity. In the present study, we identify the CLR CLEC12A as a receptor for hemozoin. Dendritic cell-T cell co-culture assays indicate that the CLEC12A/hemozoin interaction enhances CD8+ T cell cross-priming. Using the Plasmodium berghei Antwerpen-Kasapa (ANKA) mouse model of experimental cerebral malaria (ECM), we find that CLEC12A deficiency protects mice from ECM, illustrated by reduced ECM incidence and ameliorated clinical symptoms. In conclusion, we identify CLEC12A as an innate sensor of plasmodial hemozoin.


Assuntos
Hemeproteínas/imunologia , Lectinas Tipo C/imunologia , Malária Cerebral/imunologia , Plasmodium berghei/patogenicidade , Receptores Mitogênicos/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Apresentação Cruzada , Células Dendríticas/imunologia , Modelos Animais de Doenças , Granzimas/metabolismo , Imunidade Inata , Lectinas Tipo C/genética , Camundongos , Camundongos Endogâmicos C57BL , Receptores Mitogênicos/genética , Linfócitos T
12.
J Microbiol Methods ; 151: 39-43, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29857017

RESUMO

Flow cytometry enables the analysis of cells by labeling them with fluorescent probes. We describe a novel flow cytometric approach permitting reliable analysis of Coxiella (C.) burnetii-infected cells. The method quantifies infection-forming units (IFUs) in a dose-dependent manner and allows for the specific detection of infection/replication-competent coxiella in cell cultures.


Assuntos
Coxiella burnetii/isolamento & purificação , Citometria de Fluxo , Animais , Técnicas de Cultura de Células , Células Cultivadas , Contagem de Colônia Microbiana , Coxiella burnetii/metabolismo , Interações Hospedeiro-Patógeno , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA