Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Int J Med Microbiol ; 313(6): 151590, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38056089

RESUMEN

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.


Asunto(s)
Coxiella burnetii , Salud Única , Fiebre Q , Animales , Humanos , Coxiella burnetii/genética , Fiebre Q/epidemiología , Fiebre Q/prevención & control , Zoonosis/epidemiología , Zoonosis/prevención & control , Estudios Interdisciplinarios
2.
Vaccine ; 41(33): 4798-4807, 2023 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-37357077

RESUMEN

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.


Asunto(s)
Coxiella burnetii , Fiebre Q , Animales , Ovinos , Bovinos , Fiebre Q/prevención & control , Fiebre Q/veterinaria , Vacunas de Productos Inactivados , Vacunas Bacterianas , Inmunidad Celular , Vacunación/veterinaria , Vacunación/métodos , Interferón gamma , Cabras , Inmunoglobulina G , Inmunoglobulina M
3.
Viruses ; 15(4)2023 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-37112941

RESUMEN

Respiratory tract epithelium infection plays a primary role in Nipah virus (NiV) pathogenesis and transmission. Knowledge about infection dynamics and host responses to NiV infection in respiratory tract epithelia is scarce. Studies in non-differentiated primary respiratory tract cells or cell lines indicate insufficient interferon (IFN) responses. However, studies are lacking in the determination of complex host response patterns in differentiated respiratory tract epithelia for the understanding of NiV replication and spread in swine. Here we characterized infection and spread of NiV in differentiated primary porcine bronchial epithelial cells (PBEC) cultivated at the air-liquid interface (ALI). After the initial infection of only a few apical cells, lateral spread for 12 days with epithelium disruption was observed without releasing substantial amounts of infectious virus from the apical or basal sides. Deep time course proteomics revealed pronounced upregulation of genes related to type I/II IFN, immunoproteasomal subunits, transporter associated with antigen processing (TAP)-mediated peptide transport, and major histocompatibility complex (MHC) I antigen presentation. Spliceosomal factors were downregulated. We propose a model in which NiV replication in PBEC is slowed by a potent and broad type I/II IFN host response with conversion from 26S proteasomes to immunoproteasomal antigen processing and improved MHC I presentation for adaptive immunity priming. NiV induced cytopathic effects could reflect the focal release of cell-associated NiV, which may contribute to efficient airborne viral spread between pigs.


Asunto(s)
Virus Nipah , Animales , Porcinos , Virus Nipah/fisiología , Proteoma/metabolismo , Células Epiteliales , Replicación Viral , Mucosa Respiratoria , Células Cultivadas
4.
Infect Immun ; 91(2): e0032322, 2023 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-36688662

RESUMEN

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.


Asunto(s)
Coxiella burnetii , Fiebre Q , Humanos , Factor de Crecimiento Transformador beta , Fiebre Q/microbiología , Interferón gamma , Células Dendríticas
5.
Microbiol Spectr ; 10(6): e0281722, 2022 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-36219107

RESUMEN

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.


Asunto(s)
Chlamydia trachomatis , Nanotubos , Humanos , Células HEK293 , Comunicación Celular , Nanotubos/química
6.
Vet Sci ; 8(11)2021 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-34822625

RESUMEN

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.

7.
Methods Protoc ; 4(2)2021 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-33917811

RESUMEN

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.
Vaccine ; 39(10): 1499-1507, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33558108

RESUMEN

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.


Asunto(s)
Coxiella burnetii , Fiebre Q , Enfermedades de las Ovejas , Animales , Femenino , Alemania , Cabras , Inmunidad Humoral , Embarazo , Fiebre Q/prevención & control , Fiebre Q/veterinaria , Ovinos , Enfermedades de las Ovejas/prevención & control , Vacunación/veterinaria
9.
Vaccines (Basel) ; 9(1)2021 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-33466701

RESUMEN

The live genetically-engineered oral rabies virus (RABV) variant SPBN GASGAS induces long-lasting immunity in foxes and protection against challenge with an otherwise lethal dose of RABV field strains both after experimental oral and parenteral routes of administration. Induction of RABV-specific binding antibodies and immunoglobulin isotypes (IgM, total IgG, IgG1, IgG2) were comparable in orally and parenterally vaccinated foxes. Differences were only observed in the induction of virus-neutralizing (VNA) titers, which were significantly higher in the parenterally vaccinated group. The dynamics of rabies-specific antibodies pre- and post-challenge (365 days post vaccination) suggest the predominance of type-1 immunity protection of SPBN GASGAS. Independent of the route of administration, in the absence of IgG1 the immune response to SPBN GAGAS was mainly IgG2 driven. Interestingly, vaccination with SPBN GASGAS does not cause significant differences in inducible IFN-γ production in vaccinated animals, indicating a relatively weak cellular immune response during challenge. Notably, the parenteral application of SPBN GASGAS did not induce any adverse side effects in foxes, thus supporting safety studies of this oral rabies vaccine in various species.

10.
PLoS Pathog ; 16(10): e1008948, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33045019

RESUMEN

Pathogenicity often differs dramatically among even closely related arenavirus species. For instance, Junín virus (JUNV), the causative agent of Argentine hemorrhagic fever (AHF), is closely related to Tacaribe virus (TCRV), which is normally avirulent in humans. While little is known about how host cell pathways are regulated in response to arenavirus infection, or how this contributes to virulence, these two viruses have been found to differ markedly in their ability to induce apoptosis. However, details of the mechanism(s) governing the apoptotic response to arenavirus infections are unknown. Here we confirm that TCRV-induced apoptosis is mitochondria-regulated, with associated canonical hallmarks of the intrinsic apoptotic pathway, and go on to identify the pro- and anti-apoptotic Bcl-2 factors responsible for regulating this process. In particular, levels of the pro-apoptotic BH3-only proteins Noxa and Puma, as well as their canonical transcription factor p53, were strongly increased. Interestingly, TCRV infection also led to the accumulation of the inactive phosphorylated form of another pro-apoptotic BH3-only protein, Bad (i.e. as phospho-Bad). Knockout of Noxa or Puma suppressed apoptosis in response to TCRV infection, whereas silencing of Bad increased apoptosis, confirming that these factors are key regulators of apoptosis induction in response to TCRV infection. Further, we found that while the highly pathogenic JUNV does not induce caspase activation, it still activated upstream pro-apoptotic factors, consistent with current models suggesting that JUNV evades apoptosis by interfering with caspase activation through a nucleoprotein-mediated decoy function. This new mechanistic insight into the role that individual BH3-only proteins and their regulation play in controlling apoptotic fate in arenavirus-infected cells provides an important experimental framework for future studies aimed at dissecting differences in the apoptotic responses between arenaviruses, their connection to other cell signaling events and ultimately the relationship of these processes to pathogenesis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis , Infecciones por Arenaviridae/patología , Arenavirus del Nuevo Mundo/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Replicación Viral , Proteína Letal Asociada a bcl/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Infecciones por Arenaviridae/genética , Infecciones por Arenaviridae/metabolismo , Infecciones por Arenaviridae/virología , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Dominios Proteicos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteína Letal Asociada a bcl/genética
11.
Infect Immun ; 88(11)2020 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-32817330

RESUMEN

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.


Asunto(s)
Degranulación de la Célula/inmunología , Inmunidad Celular/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/microbiología , Fiebre Q/inmunología , Animales , Coxiella burnetii , Ratones
12.
Infect Immun ; 88(1)2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31658957

RESUMEN

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.


Asunto(s)
Comunicación Celular , Infecciones por Chlamydia/inmunología , Chlamydophila psittaci/inmunología , Células Dendríticas/metabolismo , Exosomas/metabolismo , Inmunidad Innata , Células Asesinas Naturales/metabolismo , Animales , Células Cultivadas , Factores Inmunológicos/metabolismo , Interferón gamma/metabolismo , Ratones , Modelos Teóricos
13.
Sci Rep ; 9(1): 4799, 2019 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-30886314

RESUMEN

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.


Asunto(s)
Chlamydophila psittaci/inmunología , Inmunidad Celular , Células Asesinas Naturales/inmunología , Psitacosis/inmunología , Células TH1/inmunología , Animales , Antígeno CD146/metabolismo , Comunicación Celular/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Interferón gamma/metabolismo , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/microbiología , Ratones , Cultivo Primario de Células , Proteína Quinasa C-theta/metabolismo , Psitacosis/sangre , Psitacosis/microbiología , Bazo/citología
14.
J Microbiol Methods ; 151: 39-43, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29857017

RESUMEN

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.


Asunto(s)
Coxiella burnetii/aislamiento & purificación , Citometría de Flujo , Animales , Técnicas de Cultivo de Célula , Células Cultivadas , Recuento de Colonia Microbiana , Coxiella burnetii/metabolismo , Interacciones Huésped-Patógeno , Ratones
15.
Int J Med Microbiol ; 308(1): 185-196, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29126745

RESUMEN

Autophagy is an evolutionarily ancient and highly conserved eukaryotic mechanism that targets cytoplasmic material for degradation. Autophagic flux involves the formation of autophagosomes and their degradation by lysosomes. The process plays a crucial role in maintaining cellular homeostasis and responds to various environmental conditions. While autophagy had previously been thought to be a non-selective process, it is now clear that it can also selectively target cellular organelles, such as mitochondria (referred to as mitophagy) and/or invading pathogens (referred to as xenophagy). Selective autophagy is characterized by specific substrate recognition and requires distinct cellular adaptor proteins. Here we review xenophagic mechanisms involved in the recognition and autolysosomal or autophagolysosomal degradation of different intracellular bacteria. In this context, we also discuss a recently discovered cellular self-defense pathway, termed mito-xenophagy, which occurs during bacterial infection of dendritic cells and depends on a TNF-α-mediated metabolic switch from oxidative phosphorylation to glycolysis.


Asunto(s)
Autofagosomas/microbiología , Autofagia , Bacterias/inmunología , Bacterias/patogenicidad , Lisosomas/microbiología , Animales , Autofagosomas/metabolismo , Citoplasma/microbiología , Células Dendríticas/metabolismo , Células Dendríticas/microbiología , Humanos , Lisosomas/metabolismo , Mitocondrias/metabolismo , Mitocondrias/microbiología
16.
Int J Cancer ; 141(12): 2505-2516, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-28857157

RESUMEN

Pancreatic cancer is the 8th most common cause of cancer-related deaths worldwide and the tumor with the poorest prognosis of all solid malignancies. In 1957, it was discovered that Newcastle disease virus (NDV) has oncolytic properties on tumor cells. To study the oncolytic properties of NDV in pancreatic cancer a single dose was administered intravenously in a syngeneic orthotopic tumor model using two different murine pancreatic adenocarcinoma cell lines (DT6606PDA, Panc02). Tumor growth was monitored and immune response was analyzed. A single treatment with NDV inhibited DT6606PDA tumor growth in mice and prevented recurrence for a period of three months. Tumor infiltration and systemic activation of NK cells, cytotoxic and helper T-cells was enhanced. NDV-induced melting of Panc02 tumors until d7 pi, but they recurred displaying unrestricted tumor growth, low immunogenicity and inhibition of tumor-specific immune response. Arrest of DT6606PDA tumor growth and rejection was mediated by activation of NK cells and a specific antitumor immune response via T-cells. Panc02 tumors rapidly decreased until d7 pi, but henceforth tumors characterized by the ability to perform immune-regulatory functions reappeared. Our results demonstrated that NDV-activated immune cells are able to reject tumors provided that an adaptive antitumor immune response can be initiated. However, activated NK cells that are abundant in Panc02 tumors lead to outgrowth of nonimmunogenic tumor cells with inhibitory properties. Our study emphasizes the importance of an adaptive immune response, which is initiated by NDV to mediate long-term tumor surveillance in addition to direct oncolysis.


Asunto(s)
Inmunidad Adaptativa , Recurrencia Local de Neoplasia/prevención & control , Virus de la Enfermedad de Newcastle/inmunología , Virus Oncolíticos/inmunología , Neoplasias Pancreáticas/inmunología , Animales , Línea Celular Tumoral , Humanos , Células Asesinas Naturales/metabolismo , Activación de Linfocitos , Ratones , Viroterapia Oncolítica , Neoplasias Pancreáticas/patología , Linfocitos T Colaboradores-Inductores/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Sci Rep ; 7(1): 3923, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28634388

RESUMEN

Chlamydiae are bacterial pathogens that grow in vacuolar inclusions. Dendritic cells (DCs) disintegrate these compartments, thereby eliminating the microbes, through auto/xenophagy, which also promotes chlamydial antigen presentation via MHC I. Here, we show that TNF-α controls this pathway by driving cytosolic phospholipase (cPLA)2-mediated arachidonic acid (AA) production. AA then impairs mitochondrial function, which disturbs the development and integrity of these energy-dependent parasitic inclusions, while a simultaneous metabolic switch towards aerobic glycolysis promotes DC survival. Tubulin deacetylase/autophagy regulator HDAC6 associates with disintegrated inclusions, thereby further disrupting their subcellular localisation and stability. Bacterial remnants are decorated with defective mitochondria, mito-aggresomal structures, and components of the ubiquitin/autophagy machinery before they are degraded via mito-xenophagy. The mechanism depends on cytoprotective HSP25/27, the E3 ubiquitin ligase Parkin and HDAC6 and promotes chlamydial antigen generation for presentation on MHC I. We propose that this novel mito-xenophagic pathway linking innate and adaptive immunity is critical for effective DC-mediated anti-bacterial resistance.


Asunto(s)
Ácido Araquidónico/metabolismo , Chlamydia/crecimiento & desarrollo , Células Dendríticas/citología , Mitofagia , Fosfolipasas A2 Citosólicas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Línea Celular , Chlamydia/citología , Técnicas de Cocultivo , Células Dendríticas/metabolismo , Células Dendríticas/microbiología , Glucólisis , Histona Desacetilasa 6/metabolismo , Ratones , Viabilidad Microbiana , Linfocitos T/citología , Linfocitos T/metabolismo
18.
FEBS Lett ; 590(21): 3920-3940, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27397851

RESUMEN

Chlamydia psittaci causes psittacosis/ornithosis in birds and is an economically important pathogen for poultry farming. It also infects nonavian domestic animals as well as rodents, and is a zoonotic human pathogen responsible for atypical pneumonia. The bacterium efficiently disseminates in host organisms causing pulmonary and systemic disease. Its rapid entry, fast replication cycle, and tight control of intracellular transport routes contribute to the host-to-host transmission and efficient growth observed with C. psittaci. Recent studies have revealed that the pathogen copes better than other chlamydial strains with proinflammatory effectors produced during the early immune reaction of infected hosts. These features likely contribute to successful infections and might explain the potent adaptation and evasion characteristics of the agent. Current findings on cell-autonomous, innate, and adaptive defenses against C. psittaci provide novel insights into the concerted immune mechanisms involved in the clearance of the pathogen. Further in-depth studies on C. psittaci and other related agents in cellular as well as animal models are needed to develop more efficient antichlamydial therapies and vaccination strategies.


Asunto(s)
Chlamydophila psittaci/fisiología , Chlamydophila psittaci/patogenicidad , Psitacosis/transmisión , Adaptación Fisiológica , Animales , Chlamydophila psittaci/genética , Genoma Bacteriano , Interacciones Huésped-Patógeno , Humanos , Evasión Inmune , Psitacosis/inmunología , Psitacosis/veterinaria
19.
Prion ; 10(3): 165-81, 2016 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-27220820

RESUMEN

Bovine spongiform encephalopathy (BSE) created a global European crisis in the 1980s and 90s, with very serious health and economic implications. Classical BSE now appears to be under control, to a great extent as a result of a global research effort that identified the sources of prions in meat and bone meal (MBM) and developed new animal-testing tools that guided policy. Priority ( www.prionpriority.eu ) was a European Union (EU) Framework Program 7 (FP7)-funded project through which 21 European research institutions and small and medium enterprises (SMEs) joined efforts between 2009 and 2014, to conduct coordinated basic and applied research on prions and prion diseases. At the end of the project, the Priority consortium drafted a position paper ( www.prionpriority.eu/Priority position paper) with its main conclusions. In the present opinion paper, we summarize these conclusions. With respect to the issue of re-introducing ruminant protein into the feed-chain, our opinion is that sustaining an absolute ban on feeding ruminant protein to ruminants is essential. In particular, the spread and impact of non-classical forms of scrapie and BSE in ruminants is not fully understood and the risks cannot be estimated. Atypical prion agents will probably continue to represent the dominant form of prion diseases in the near future in Europe. Atypical L-type BSE has clear zoonotic potential, as demonstrated in experimental models. Similarly, there are now data indicating that the atypical scrapie agent can cross various species barriers. More epidemiological data from large cohorts are necessary to reach any conclusion on the impact of its transmissibility on public health. Re-evaluations of safety precautions may become necessary depending on the outcome of these studies. Intensified searching for molecular determinants of the species barrier is recommended, since this barrier is key for important policy areas and risk assessment. Understanding the structural basis for strains and the basis for adaptation of a strain to a new host will require continued fundamental research, also needed to understand mechanisms of prion transmission, replication and how they cause nervous system dysfunction and death. Early detection of prion infection, ideally at a preclinical stage, also remains crucial for development of effective treatment strategies.


Asunto(s)
Cadena Alimentaria , Enfermedades por Prión/epidemiología , Enfermedades por Prión/prevención & control , Priones/análisis , Alimentación Animal/efectos adversos , Animales , Bovinos , Diagnóstico Precoz , Encefalopatía Espongiforme Bovina/diagnóstico , Encefalopatía Espongiforme Bovina/epidemiología , Encefalopatía Espongiforme Bovina/prevención & control , Encefalopatía Espongiforme Bovina/transmisión , Europa (Continente)/epidemiología , Humanos , Enfermedades por Prión/diagnóstico , Enfermedades por Prión/transmisión , Priones/aislamiento & purificación , Priones/metabolismo , Priones/patogenicidad , Scrapie/diagnóstico , Scrapie/epidemiología , Scrapie/prevención & control , Scrapie/transmisión
20.
J Immunol ; 195(11): 5482-94, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26519531

RESUMEN

The transporter associated with Ag processing (TAP) translocates proteasomally derived cytosolic peptides into the endoplasmic reticulum. TAP is a central component of the peptide-loading complex (PLC), to which tapasin (TPN) recruits MHC class I (MHC I) and accessory chaperones. The PLC functions to facilitate and optimize MHC I-mediated Ag presentation. The heterodimeric peptide transporter consists of two homologous subunits, TAP1 and TAP2, each of which contains an N-terminal domain (N-domain) in addition to a conserved transmembrane (TM) core segment. Each N-domain binds to the TM region of a single TPN molecule, which recruits one MHC I molecule to TAP1 and/or TAP2. Although both N-domains act as TPN-docking sites, various studies suggest a functional asymmetry within the PLC resulting in greater significance of the TAP2/TPN interaction for MHC loading. In this study, we demonstrate that the leucine-rich hydrophobic sequence stretches (with the central leucine residues L20 and L66) in the first and second TM helix of TAP2 form a functional unit acting as a docking site for optimal TPN/MHC I recruitment, whereas three distinct highly conserved arginine and/or aspartate residues inside or flanking these TM helices are dispensable. Moreover, we show that the physical interaction between TAP2 and TPN is disrupted by benzene, a compound known to interfere with hydrophobic interactions, such as those between pairing leucine zippers. No such effects were observed for the TAP1/TAP2 interaction or the complex formation between TPN and MHC I. We propose that TAP/TPN complex formation is driven by hydrophobic interactions via leucine zipper-like motifs.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Complejos Multiproteicos/ultraestructura , Miembro 3 de la Subfamilia B de Transportadores de Casetes de Unión a ATP , Transportadoras de Casetes de Unión a ATP/ultraestructura , Benceno/química , Sitios de Unión/inmunología , Transporte Biológico/inmunología , Línea Celular , Retículo Endoplásmico/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Leucina Zippers/efectos de los fármacos , Leucina Zippers/genética , Proteínas de Transporte de Membrana/ultraestructura , Complejos Multiproteicos/efectos de los fármacos , Complejos Multiproteicos/metabolismo , Unión Proteica/inmunología , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA