Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Pathog Dis ; 822024 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-39138067

RESUMO

Coxiella burnetii is a globally distributed obligate intracellular pathogen. Although often asymptomatic, infections can cause acute Q fever with influenza-like symptoms and/or severe chronic Q fever. Coxiella burnetii develops a unique replicative niche within host cells called the Coxiella-containing vacuole (CCV), facilitated by the Dot/Icm type IV secretion system translocating a cohort of bacterial effector proteins into the host. The role of some effectors has been elucidated; however, the actions of the majority remain enigmatic and the list of true effectors is disputable. This study examined CBU2016, a unique C. burnetii protein previously designated as an effector with a role in infection. We were unable to validate CBU2016 as a translocated effector protein. Employing targeted knock-out and complemented strains, we found that the loss of CBU2016 did not cause a replication defect within Hela, THP-1, J774, or iBMDM cells or in axenic media, nor did it affect the pathogenicity of C. burnetii in the Galleria mellonella infection model. The absence of CBU2016 did, however, result in a consistent decrease in the size of CCVs in HeLa cells. These results suggest that although CBU2016 may not be a Dot/Icm effector, it is still able to influence the host environment during infection.


Assuntos
Proteínas de Bactérias , Coxiella burnetii , Febre Q , Vacúolos , Coxiella burnetii/genética , Coxiella burnetii/metabolismo , Coxiella burnetii/patogenicidade , Humanos , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Vacúolos/microbiologia , Vacúolos/metabolismo , Animais , Febre Q/microbiologia , Células HeLa , Linhagem Celular , Fatores de Virulência/metabolismo , Fatores de Virulência/genética , Técnicas de Inativação de Genes , Mariposas/microbiologia , Interações Hospedeiro-Patógeno , Células THP-1
2.
Front Cell Infect Microbiol ; 14: 1394019, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38841112

RESUMO

Introduction: Coxiella burnetii is a gram-negative obligate intracellular bacterium and a zoonotic pathogen that causes human Q fever. The lack of effective antibiotics and a licensed vaccine for Coxiella in the U.S. warrants further research into Coxiella pathogenesis. Within the host cells, Coxiella replicates in an acidic phagolysosome-like vacuole termed Coxiella-containing vacuole (CCV). Previously, we have shown that the CCV pH is critical for Coxiella survival and that the Coxiella Type 4B secretion system regulates CCV pH by inhibiting the host endosomal maturation pathway. However, the trafficking pattern of the 'immature' endosomes in Coxiella- infected cells remained unclear. Methods: We transfected HeLa cells with GFP-tagged Rab proteins and subsequently infected them with mCherry-Coxiella to visualize Rab protein localization. Infected cells were immunostained with anti-Rab antibodies to confirm the Rab localization to the CCV, to quantitate Rab11a and Rab35- positive CCVs, and to quantitate total recycling endosome content of infected cells. A dual-hit siRNA mediated knockdown combined with either immunofluorescent assay or an agarose-based colony-forming unit assay were used to measure the effects of Rab11a and Rab35 knockdown on CCV area and Coxiella intracellular growth. Results: The CCV localization screen with host Rab proteins revealed that recycling endosome-associated proteins Rab11a and Rab35 localize to the CCV during infection, suggesting that CCV interacts with host recycling endosomes during maturation. Interestingly, only a subset of CCVs were Rab11a or Rab35-positive at any given time point. Quantitation of Rab11a/Rab35-positive CCVs revealed that while Rab11a interacts with the CCV more at 3 dpi, Rab35 is significantly more prevalent at CCVs at 6 dpi, suggesting that the CCV preferentially interacts with Rab11a and Rab35 depending on the stage of infection. Furthermore, we observed a significant increase in Rab11a and Rab35 fluorescent intensity in Coxiella-infected cells compared to mock, suggesting that Coxiella increases the recycling endosome content in infected cells. Finally, siRNA-mediated knockdown of Rab11a and Rab35 resulted in significantly smaller CCVs and reduced Coxiella intracellular growth, suggesting that recycling endosomal Rab proteins are essential for CCV expansion and bacterial multiplication. Discussion: Our data, for the first time, show that the CCV dynamically interacts with host recycling endosomes for Coxiella intracellular survival and potentially uncovers novel host cell factors essential for Coxiella pathogenesis.


Assuntos
Coxiella burnetii , Endossomos , Interações Hospedeiro-Patógeno , Vacúolos , Proteínas rab de Ligação ao GTP , Coxiella burnetii/metabolismo , Coxiella burnetii/crescimento & desenvolvimento , Coxiella burnetii/genética , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab de Ligação ao GTP/genética , Humanos , Vacúolos/metabolismo , Vacúolos/microbiologia , Células HeLa , Endossomos/metabolismo , Endossomos/microbiologia , Febre Q/microbiologia , Febre Q/metabolismo
3.
mBio ; 15(4): e0029924, 2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38415594

RESUMO

Many intracellular pathogens structurally disrupt the Golgi apparatus as an evolutionarily conserved promicrobial strategy. Yet, the host factors and signaling processes involved are often poorly understood, particularly for Anaplasma phagocytophilum, the agent of human granulocytic anaplasmosis. We found that A. phagocytophilum elevated cellular levels of the bioactive sphingolipid, ceramide-1-phosphate (C1P), to promote Golgi fragmentation that enables bacterial proliferation, conversion from its non-infectious to infectious form, and productive infection. A. phagocytophilum poorly infected mice deficient in ceramide kinase, the Golgi-localized enzyme responsible for C1P biosynthesis. C1P regulated Golgi morphology via activation of a PKCα/Cdc42/JNK signaling axis that culminates in phosphorylation of Golgi structural proteins, GRASP55 and GRASP65. siRNA-mediated depletion of Cdc42 blocked A. phagocytophilum from altering Golgi morphology, which impaired anterograde trafficking of trans-Golgi vesicles into and maturation of the pathogen-occupied vacuole. Cells overexpressing phosphorylation-resistant versions of GRASP55 and GRASP65 presented with suppressed C1P- and A. phagocytophilum-induced Golgi fragmentation and poorly supported infection by the bacterium. By studying A. phagocytophilum, we identify C1P as a regulator of Golgi structure and a host factor that is relevant to disease progression associated with Golgi fragmentation.IMPORTANCECeramide-1-phosphate (C1P), a bioactive sphingolipid that regulates diverse processes vital to mammalian physiology, is linked to disease states such as cancer, inflammation, and wound healing. By studying the obligate intracellular bacterium Anaplasma phagocytophilum, we discovered that C1P is a major regulator of Golgi morphology. A. phagocytophilum elevated C1P levels to induce signaling events that promote Golgi fragmentation and increase vesicular traffic into the pathogen-occupied vacuole that the bacterium parasitizes. As several intracellular microbial pathogens destabilize the Golgi to drive their infection cycles and changes in Golgi morphology is also linked to cancer and neurodegenerative disorder progression, this study identifies C1P as a potential broad-spectrum therapeutic target for infectious and non-infectious diseases.


Assuntos
Anaplasma phagocytophilum , Neoplasias , Animais , Humanos , Camundongos , Anaplasma phagocytophilum/metabolismo , Complexo de Golgi/metabolismo , Ceramidas , Mamíferos/metabolismo
4.
Pathog Dis ; 79(9)2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34755855

RESUMO

Mammals have evolved sophisticated host cell death signaling pathways as an important immune mechanism to recognize and eliminate cell intruders before they establish their replicative niche. However, intracellular bacterial pathogens that have co-evolved with their host have developed a multitude of tactics to counteract this defense strategy to facilitate their survival and replication. This requires manipulation of pro-death and pro-survival host signaling pathways during infection. Obligate intracellular bacterial pathogens are organisms that absolutely require an eukaryotic host to survive and replicate, and therefore they have developed virulence factors to prevent diverse forms of host cell death and conserve their replicative niche. This review encapsulates our current understanding of these host-pathogen interactions by exploring the most relevant findings of Anaplasma spp., Chlamydia spp., Rickettsia spp. and Coxiella burnetii modulating host cell death pathways. A detailed comprehension of the molecular mechanisms through which these obligate intracellular pathogens manipulate regulated host cell death will not only increase the current understanding of these difficult-to-study pathogens but also provide insights into new tools to study regulated cell death and the development of new therapeutic approaches to control infection.


Assuntos
Fenômenos Fisiológicos Bacterianos , Suscetibilidade a Doenças , Interações Hospedeiro-Patógeno , Animais , Biomarcadores , Morte Celular/imunologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Espaço Intracelular/imunologia , Espaço Intracelular/metabolismo , Espaço Intracelular/microbiologia , Lisossomos/imunologia , Lisossomos/metabolismo , Lisossomos/microbiologia , Viabilidade Microbiana/imunologia , Estresse Oxidativo , Fagocitose , Especificidade da Espécie , Fatores de Virulência
5.
Biochem J ; 476(19): 2851-2867, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31527117

RESUMO

Coxiella burnetii is a Gram-negative bacterium which causes Q fever, a complex and life-threatening infection with both acute and chronic presentations. C. burnetii invades a variety of host cell types and replicates within a unique vacuole derived from the host cell lysosome. In order to understand how C. burnetii survives within this intracellular niche, we have investigated the carbon metabolism of both intracellular and axenically cultivated bacteria. Both bacterial populations were shown to assimilate exogenous [13C]glucose or [13C]glutamate, with concomitant labeling of intermediates in glycolysis and gluconeogenesis, and in the TCA cycle. Significantly, the two populations displayed metabolic pathway profiles reflective of the nutrient availabilities within their propagated environments. Disruption of the C. burnetii glucose transporter, CBU0265, by transposon mutagenesis led to a significant decrease in [13C]glucose utilization but did not abolish glucose usage, suggesting that C. burnetii express additional hexose transporters which may be able to compensate for the loss of CBU0265. This was supported by intracellular infection of human cells and in vivo studies in the insect model showing loss of CBU0265 had no impact on intracellular replication or virulence. Using this mutagenesis and [13C]glucose labeling approach, we identified a second glucose transporter, CBU0347, the disruption of which also showed significant decreases in 13C-label incorporation but did not impact intracellular replication or virulence. Together, these analyses indicate that C. burnetii may use multiple carbon sources in vivo and exhibits greater metabolic flexibility than expected.


Assuntos
Coxiella burnetii/metabolismo , Glucose/metabolismo , Ácido Glutâmico/metabolismo , Interações Hospedeiro-Patógeno , Febre Q/microbiologia , Virulência/fisiologia , Animais , Proteínas de Bactérias/metabolismo , Transporte Biológico , Coxiella burnetii/patogenicidade , Gluconeogênese/fisiologia , Glicólise/fisiologia , Células HeLa , Humanos , Lepidópteros/microbiologia , Proteínas de Membrana Transportadoras/metabolismo , Células THP-1
6.
Autophagy ; 14(10): 1710-1725, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29973118

RESUMO

Coxiella burnetii is an intracellular bacterial pathogen which causes Q fever, a human infection with the ability to cause chronic disease with potentially life-threatening outcomes. In humans, Coxiella infects alveolar macrophages where it replicates to high numbers in a unique, pathogen-directed lysosome-derived vacuole. This compartment, termed the Coxiella-containing vacuole (CCV), has a low internal pH and contains markers both of lysosomes and autophagosomes. The CCV membrane is also enriched with CLTC (clathrin heavy chain) and this contributes to the success of the CCV. Here, we describe a role for CLTC, a scaffolding protein of clathrin-coated vesicles, in facilitating the fusion of autophagosomes with the CCV. During gene silencing of CLTC, CCVs are unable to fuse with each other, a phenotype also seen when silencing genes involved in macroautophagy/autophagy. MAP1LC3B/LC3B, which is normally observed inside the CCV, is excluded from CCVs in the absence of CLTC. Additionally, this study demonstrates that autophagosome fusion contributes to CCV size as cell starvation and subsequent autophagy induction leads to further CCV expansion. This is CLTC dependent, as the absence of CLTC renders autophagosomes no longer able to contribute to the expansion of the CCV. This investigation provides a functional link between CLTC and autophagy in the context of Coxiella infection and highlights the CCV as an important tool to explore the interactions between these vesicular trafficking pathways.


Assuntos
Autofagia , Cadeias Pesadas de Clatrina/metabolismo , Coxiella/metabolismo , Vacúolos/metabolismo , Autofagossomos/metabolismo , Células HeLa , Humanos , Membranas Intracelulares/metabolismo , Fusão de Membrana , Proteínas Associadas aos Microtúbulos/metabolismo , Fenótipo
7.
J Proteome Res ; 15(12): 4675-4685, 2016 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-27934296

RESUMO

Intracellular actin-based motility of the melioidosis pathogen Burkholderia pseudomallei requires the bacterial factor BimA. Located at one pole of the bacterium, BimA recruits and polymerizes cellular actin to promote bacterial motility within and between cells. Here, we describe an affinity approach coupled with mass spectrometry to identify cellular proteins recruited to BimA-expressing bacteria under conditions that promote actin polymerization. We identified a group of cellular proteins that are recruited to the B. pseudomallei surface in a BimA-dependent manner, a subset of which were independently validated with specific antisera including the ubiquitous scaffold protein Ras GTPase-activating-like protein (IQGAP1). IQGAP1 integrates several key cellular signaling pathways including those involved in actin dynamics and has been shown to be involved in the adhesion of attaching and effacing Escherichia coli to infected cells and invasion of host cells by Salmonella enterica serovar Typhimurium. Although a direct interaction between BimA and IQGAP1 could not be detected using either conventional pulldown or yeast two hybrid techniques, confocal microscopy revealed that IQGAP1 is recruited to B. pseudomallei actin tails in infected cells, and siRNA-mediated knockdown highlighted a role for this protein in controlling the length and actin density of B. pseudomallei actin tails.


Assuntos
Actinas/metabolismo , Burkholderia pseudomallei/química , Movimento Celular , Proteínas de Bactérias/análise , Proteínas de Bactérias/fisiologia , Burkholderia pseudomallei/citologia , Polaridade Celular , Humanos , Proteínas dos Microfilamentos/metabolismo , Proteínas dos Microfilamentos/fisiologia , Polimerização , Proteínas Ativadoras de ras GTPase/metabolismo , Proteínas Ativadoras de ras GTPase/fisiologia
8.
Virulence ; 6(5): 412-3, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26126122

RESUMO

In this issue of Virulence, Ramon-Garcia et al. demonstrate the requirement of a mycobacterial efflux pump during growth on cholesterol. In this editorial I replace the study in the context of nutrient acquisition by Mycobacterium tuberculosis.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Colesterol/metabolismo , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Mycobacterium bovis/crescimento & desenvolvimento , Mycobacterium bovis/genética
9.
J Biol Chem ; 290(13): 8383-95, 2015 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-25635050

RESUMO

Bacterial pathogens often interfere with host tyrosine phosphorylation cascades to control host responses and cause infection. Given the role of tyrosine phosphorylation events in different human infections and our previous results showing the activation of the tyrosine kinase Src upon incubation of cells with Listeria monocytogenes, we searched for novel host proteins undergoing tyrosine phosphorylation upon L. monocytogenes infection. We identify the heavy chain of the non-muscle myosin IIA (NMHC-IIA) as being phosphorylated in a specific tyrosine residue in response to L. monocytogenes infection. We characterize this novel post-translational modification event and show that, upon L. monocytogenes infection, Src phosphorylates NMHC-IIA in a previously uncharacterized tyrosine residue (Tyr-158) located in its motor domain near the ATP-binding site. In addition, we found that other intracellular and extracellular bacterial pathogens trigger NMHC-IIA tyrosine phosphorylation. We demonstrate that NMHC-IIA limits intracellular levels of L. monocytogenes, and this is dependent on the phosphorylation of Tyr-158. Our data suggest a novel mechanism of regulation of NMHC-IIA activity relying on the phosphorylation of Tyr-158 by Src.


Assuntos
Listeria monocytogenes/fisiologia , Listeriose/enzimologia , Miosina não Muscular Tipo IIA/metabolismo , Processamento de Proteína Pós-Traducional , Quinases da Família src/metabolismo , Sequência de Aminoácidos , Carga Bacteriana , Células CACO-2 , Ativação Enzimática , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Listeriose/microbiologia , Fosforilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA