Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Infect Immun ; 92(7): e0005324, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38837340

RESUMEN

Coxiella burnetii is an obligate intracellular bacteria that causes the global zoonotic disease Q Fever. Treatment options for chronic infection are limited, and the development of novel therapeutic strategies requires a greater understanding of how C. burnetii interacts with immune signaling. Cell death responses are known to be manipulated by C. burnetii, but the role of caspase-8, a central regulator of multiple cell death pathways, has not been investigated. In this research, we studied bacterial manipulation of caspase-8 signaling and the significance of caspase-8 to C. burnetii infection, examining bacterial replication, cell death induction, and cytokine signaling. We measured caspase, RIPK, and MLKL activation in C. burnetii-infected tumor necrosis factor alpha (TNFα)/cycloheximide-treated THP-1 macrophage-like cells and TNFα/ZVAD-treated L929 cells to assess apoptosis and necroptosis signaling. Additionally, we measured C. burnetii replication, cell death, and TNFα induction over 12 days in RIPK1-kinase-dead, RIPK3-kinase-dead, or RIPK3-kinase-dead-caspase-8-/- bone marrow-derived macrophages (BMDMs) to understand the significance of caspase-8 and RIPK1/3 during infection. We found that caspase-8 is inhibited by C. burnetii, coinciding with inhibition of apoptosis and increased susceptibility to necroptosis. Furthermore, C. burnetii replication was increased in BMDMs lacking caspase-8, but not in those lacking RIPK1/3 kinase activity, corresponding with decreased TNFα production and reduced cell death. As TNFα is associated with the control of C. burnetii, this lack of a TNFα response may allow for the unchecked bacterial growth we saw in caspase-8-/- BMDMs. This research identifies and explores caspase-8 as a key regulator of C. burnetii infection, opening novel therapeutic doors.


Asunto(s)
Caspasa 8 , Coxiella burnetii , Macrófagos , Fiebre Q , Factor de Necrosis Tumoral alfa , Caspasa 8/metabolismo , Animales , Factor de Necrosis Tumoral alfa/metabolismo , Macrófagos/microbiología , Macrófagos/metabolismo , Macrófagos/inmunología , Ratones , Fiebre Q/microbiología , Fiebre Q/inmunología , Fiebre Q/metabolismo , Humanos , Apoptosis , Transducción de Señal , Línea Celular , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Células THP-1
2.
Infect Immun ; 91(1): e0019922, 2023 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-36537791

RESUMEN

MicroRNAs (miRNAs), a class of small noncoding RNAs, are critical to gene regulation in eukaryotes. They are involved in modulating a variety of physiological processes, including the host response to intracellular infections. Little is known about miRNA functions during infection by Coxiella burnetii, the causative agent of human Q fever. This bacterial pathogen establishes a large replicative vacuole within macrophages by manipulating host processes such as apoptosis and autophagy. We investigated miRNA expression in C. burnetii-infected macrophages and identified several miRNAs that were down- or upregulated during infection. We further explored the functions of miR-143-3p, an miRNA whose expression is downregulated in macrophages infected with C. burnetii, and show that increasing the abundance of this miRNA in human cells results in increased apoptosis and reduced autophagy-conditions that are unfavorable to C. burnetii intracellular growth. In sum, this study demonstrates that C. burnetii infection elicits a robust miRNA-based host response, and because miR-143-3p promotes apoptosis and inhibits autophagy, downregulation of miR-143-3p expression during C. burnetii infection likely benefits the pathogen.


Asunto(s)
Coxiella burnetii , MicroARNs , Fiebre Q , Humanos , Coxiella burnetii/fisiología , MicroARNs/genética , MicroARNs/metabolismo , Interacciones Huésped-Patógeno/genética , Fiebre Q/genética , Fiebre Q/metabolismo , Macrófagos/microbiología , Vacuolas/microbiología
3.
Microbiol Spectr ; 10(4): e0248421, 2022 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-35913176

RESUMEN

Coxiella burnetii requires a type IVB secretion system (T4SS) to promote intracellular replication and virulence. We hypothesized that Coxiella employs its T4SS to secrete effectors that enable stealthy colonization of immune cells. To address this, we used RNA sequencing to compare the transcriptional response of murine bone marrow-derived macrophages (BMDM) infected with those of wild-type Coxiella and a T4SS-null mutant at 8 and 24 h postinfection. We found a T4SS-independent upregulation of proinflammatory transcripts which was consistent with a proinflammatory polarization phenotype. Despite this, infected BMDM failed to completely polarize, as evidenced by modest surface expression of CD38 and CD11c, nitrate production, and reduced proinflammatory cytokine and chemokine secretion compared to positive controls. As these BMDM permitted replication of C. burnetii, we employed them to identify T4SS effectors that are essential in the specific cellular context of a primary macrophage. We found five Himar1 transposon mutants in T4SS effectors that had a replication defect in BMDM but not J774A.1 cells. The mutants were also attenuated in a SCID mouse model of infection. Among these candidate virulence factors, we found that CBU1639 contributed to the inhibition of macrophage proinflammatory responses to Coxiella infection. These data demonstrate that while T4SS is dispensable for the stealthy invasion of primary macrophages, Coxiella has evolved multiple T4SS effectors that specifically target macrophage function to proliferate within that specific cellular context. IMPORTANCE Coxiella burnetii, the causative agent of Q fever, preferentially infects macrophages of the respiratory tract when causing human disease. This work describes how primary macrophages respond to C. burnetii at the earliest stages of infection, before bacterial replication. We found that while infected macrophages increase expression of proinflammatory genes after bacterial entry, they fail to activate the accompanying antibacterial functions that might ultimately control the infection. This disconnect between initial response and downstream function was not mediated by the bacterium's type IVB secretion system, suggesting that Coxiella has other virulence factors that dampen host responses early in the infection process. Nevertheless, we were able to identify several type IVB secreted effectors that were specifically required for survival in macrophages and mice. This work is the first to identify type IVB secretion effectors that are specifically required for infection and replication within primary macrophages.


Asunto(s)
Coxiella burnetii , Fiebre Q , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Coxiella burnetii/genética , Interacciones Huésped-Patógeno/fisiología , Humanos , Macrófagos/microbiología , Ratones , Ratones SCID , Fiebre Q/metabolismo , Fiebre Q/microbiología , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
4.
Elife ; 112022 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-35137689

RESUMEN

Background: Chronic Q fever is a zoonosis caused by the bacterium Coxiella burnetii which can manifest as infection of an abdominal aortic aneurysm (AAA). Antibiotic therapy often fails, resulting in severe morbidity and high mortality. Whereas previous studies have focused on inflammatory processes in blood, the aim of this study was to investigate local inflammation in aortic tissue. Methods: Multiplex immunohistochemistry was used to investigate local inflammation in Q fever AAAs compared to atherosclerotic AAAs in aorta tissue specimen. Two six-plex panels were used to study both the innate and adaptive immune systems. Results: Q fever AAAs and atherosclerotic AAAs contained similar numbers of CD68+ macrophages and CD3+ T cells. However, in Q fever AAAs, the number of CD68+CD206+ M2 macrophages was increased, while expression of GM-CSF was decreased compared to atherosclerotic AAAs. Furthermore, Q fever AAAs showed an increase in both the number of CD8+ cytotoxic T cells and CD3+CD8-FoxP3+ regulatory T cells. Finally, Q fever AAAs did not contain any well-defined granulomas. Conclusions: These findings demonstrate that despite the presence of pro-inflammatory effector cells, persistent local infection with C. burnetii is associated with an immune-suppressed microenvironment. Funding: This work was supported by SCAN consortium: European Research Area - CardioVascualar Diseases (ERA-CVD) grant [JTC2017-044] and TTW-NWO open technology grant [STW-14716].


Asunto(s)
Inmunidad Adaptativa/inmunología , Aneurisma de la Aorta Abdominal/inmunología , Aterosclerosis/inmunología , Inmunidad Innata/inmunología , Fiebre Q/inmunología , Anciano , Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/microbiología , Aterosclerosis/metabolismo , Aterosclerosis/microbiología , Femenino , Humanos , Inmunohistoquímica/métodos , Inflamación/inmunología , Inflamación/microbiología , Macrófagos/metabolismo , Masculino , Persona de Mediana Edad , Fiebre Q/metabolismo , Fiebre Q/microbiología , Linfocitos T/metabolismo
5.
J Biol Chem ; 295(21): 7391-7403, 2020 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-32303638

RESUMEN

The intracellular bacterial pathogen Coxiella burnetii is the etiological agent of the emerging zoonosis Q fever. Crucial to its pathogenesis is type 4b secretion system-mediated secretion of bacterial effectors into host cells that subvert host cell membrane trafficking, leading to the biogenesis of a parasitophorous vacuole for intracellular replication. The characterization of prokaryotic serine/threonine protein kinases in bacterial pathogens is emerging as an important strategy to better understand host-pathogen interactions. In this study, we investigated CstK (for Coxiella Ser/Thr kinase), a protein kinase identified in C. burnetii by in silico analysis. We demonstrate that this putative protein kinase undergoes autophosphorylation on Thr and Tyr residues and phosphorylates a classical eukaryotic protein kinase substrate in vitro This dual Thr-Tyr kinase activity is also observed for a eukaryotic dual-specificity Tyr phosphorylation-regulated kinase class. We found that CstK is translocated during infections and localizes to Coxiella-containing vacuoles (CCVs). Moreover, a CstK-overexpressing C. burnetii strain displayed a severe CCV development phenotype, suggesting that CstK fine-tunes CCV biogenesis during the infection. Protein-protein interaction experiments identified the Rab7 GTPase-activating protein TBC1D5 as a candidate CstK-specific target, suggesting a role for this host GTPase-activating protein in Coxiella infections. Indeed, CstK co-localized with TBC1D5 in noninfected cells, and TBC1D5 was recruited to CCVs in infected cells. Accordingly, TBC1D5 depletion from infected cells significantly affected CCV development. Our results indicate that CstK functions as a bacterial effector protein that interacts with the host protein TBC1D5 during vacuole biogenesis and intracellular replication.


Asunto(s)
Proteínas Bacterianas/metabolismo , Coxiella burnetii/enzimología , Proteínas Activadoras de GTPasa/metabolismo , Proteínas Quinasas/metabolismo , Fiebre Q/metabolismo , Vacuolas/metabolismo , Proteínas Bacterianas/genética , Línea Celular Tumoral , Coxiella burnetii/genética , Proteínas Activadoras de GTPasa/genética , Humanos , Fosforilación , Proteínas Quinasas/genética , Fiebre Q/genética , Vacuolas/genética , Vacuolas/microbiología
6.
Infect Immun ; 88(7)2020 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-32284364

RESUMEN

Coxiella burnetii is the causative agent of human Q fever, eliciting symptoms that range from acute fever and fatigue to chronic fatal endocarditis. C. burnetii is a Gram-negative intracellular bacterium that replicates within an acidic lysosome-like parasitophorous vacuole (PV) in human macrophages. During intracellular growth, C. burnetii delivers bacterial proteins directly into the host cytoplasm using a Dot/Icm type IV secretion system (T4SS). Multiple T4SS effectors localize to and/or disrupt the endoplasmic reticulum (ER) and secretory transport, but their role in infection is unknown. During microbial infection, unfolded nascent proteins may exceed the folding capacity of the ER, activating the unfolded protein response (UPR) and restoring the ER to its normal physiological state. A subset of intracellular pathogens manipulates the UPR to promote survival and replication in host cells. In this study, we investigated the impact of C. burnetii infection on activation of the three arms of the UPR. An inhibitor of the UPR antagonized PV expansion in macrophages, indicating this process is needed for bacterial replication niche formation. Protein kinase RNA-like ER kinase (PERK) signaling was activated during infection, leading to increased levels of phosphorylated eukaryotic initiation factor α, which was required for C. burnetii growth. Increased production and nuclear translocation of the transcription factor ATF4 also occurred, which normally drives expression of the proapoptotic C/EBP homologous protein (CHOP). CHOP protein production increased during infection; however, C. burnetii actively prevented CHOP nuclear translocation and downstream apoptosis in a T4SS-dependent manner. The results collectively demonstrate interplay between C. burnetii and specific components of the eIF2α signaling cascade to parasitize human macrophages.


Asunto(s)
Coxiella burnetii/fisiología , Factor 2 Eucariótico de Iniciación/metabolismo , Interacciones Huésped-Patógeno , Fiebre Q/metabolismo , Fiebre Q/microbiología , Factor de Transcripción Activador 6/metabolismo , Sistemas de Secreción Bacterianos , Histonas/metabolismo , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Transporte de Proteínas , Factor de Transcripción CHOP/metabolismo
7.
Cell Microbiol ; 22(5): e13154, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31872956

RESUMEN

Coxiella burnetii, the causative agent of the zoonotic disease Q fever, is a Gram-negative bacterium that replicates inside macrophages within a highly oxidative vacuole. Screening of a transposon mutant library suggested that sdrA, which encodes a putative short-chain dehydrogenase, is required for intracellular replication. Short-chain dehydrogenases are NADP(H)-dependent oxidoreductases, and SdrA contains a predicted NADP+ binding site, suggesting it may facilitate NADP(H) regeneration by C. burnetii, a key process for surviving oxidative stress. Purified recombinant 6×His-SdrA was able to convert NADP+ to NADP(H) in vitro. Mutation to alanine of a conserved glycine residue at position 12 within the predicted NADP binding site abolished significant enzymatic activity. Complementation of the sdrA mutant (sdrA::Tn) with plasmid-expressed SdrA restored intracellular replication to wild-type levels, but expressing enzymatically inactive G12A_SdrA did not. The sdrA::Tn mutant was more susceptible in vitro to oxidative stress, and treating infected host cells with L-ascorbate, an anti-oxidant, partially rescued the intracellular growth defect of sdrA::Tn. Finally, stable isotope labelling studies demonstrated a shift in flux through metabolic pathways in sdrA::Tn consistent with the presence of increased oxidative stress, and host cells infected with sdrA::Tn had elevated levels of reactive oxygen species compared with C. burnetii NMII.


Asunto(s)
Coxiella burnetii/metabolismo , NADP/metabolismo , Estrés Oxidativo , Coxiella burnetii/crecimiento & desarrollo , Citoplasma/metabolismo , Células HeLa , Humanos , Macrófagos/microbiología , Mutación , NADP/genética , Fiebre Q/metabolismo , Fiebre Q/microbiología , Regeneración , Vacuolas/microbiología
8.
J Transl Med ; 17(1): 157, 2019 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-31088495

RESUMEN

BACKGROUND: Q fever fatigue syndrome (QFS) is a well-documented state of prolonged fatigue following around 20% of acute Q fever infections. It has been hypothesized that low grade inflammation plays a role in its aetiology. In this study, we aimed to identify transcriptome profiles that could aid to better understand the pathophysiology of QFS. METHODS: RNA of monocytes was collected from QFS patients (n = 10), chronic fatigue syndrome patients (CFS, n = 10), Q fever seropositive controls (n = 10), and healthy controls (n = 10) who were age- (± 5 years) and sex-matched. Transcriptome analysis was performed using RNA sequencing. RESULTS: Mitochondrial-derived peptide (MDP)-coding genes MT-RNR2 (humanin) and MT-RNR1 (MOTS-c) were differentially expressed when comparing QFS (- 4.8 log2-fold-change P = 2.19 × 10-9 and - 4.9 log2-fold-change P = 4.69 × 10-8), CFS (- 5.2 log2-fold-change, P = 3.49 × 10-11 - 4.4 log2-fold-change, P = 2.71 × 10-9), and Q fever seropositive control (- 3.7 log2-fold-change P = 1.78 × 10-6 and - 3.2 log2-fold-change P = 1.12 × 10-5) groups with healthy controls, resulting in a decreased median production of humanin in QFS patients (371 pg/mL; Interquartile range, IQR, 325-384), CFS patients (364 pg/mL; IQR 316-387), and asymptomatic Q fever seropositive controls (354 pg/mL; 292-393). CONCLUSIONS: Expression of MDP-coding genes MT-RNR1 (MOTS-c) and MT-RNR2 (humanin) is decreased in CFS, QFS, and, to a lesser extent, in Q fever seropositive controls, resulting in a decreased production of humanin. These novel peptides might indeed be important in the pathophysiology of both QFS and CFS.


Asunto(s)
Síndrome de Fatiga Crónica/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Fiebre Q/metabolismo , Adulto , Síndrome de Fatiga Crónica/genética , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Análisis de Componente Principal , Fiebre Q/genética
9.
PLoS One ; 14(2): e0212202, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30763357

RESUMEN

Cyclic Adenosine 3',5'-monophosphate (cAMP) is a key second messenger known to directly regulate not only the protein kinase A (PKA) activity but also other important molecules such as the exchange protein activated by cAMP (EPAC), which is as a guanine nucleotide exchange factor (GEF) of the low molecular weight GTPase, Rap2. Coxiella burnetii is a Gram negative bacterium that survives and grows in a large Coxiella replicative vacuole (CRV), which displays lysosomal and autophagic features. In this report, we present evidence that both, EPAC and its downstream effector Rap2b, were recruited to the CRV. The transient over-expression of the Rap2b wt protein, but not its inactive mutant Rap2b ΔAAX, markedly inhibited the development of the large CRV. Additionally, Rap2b wtinhibited the fusion of early Coxiella phagosomes with the fully developed CRV, indicating that homotypic fusion events are altered in the presence of high levels of Rap2b wt. Likewise, the fusion of endosome/lysosomal compartments (heterotypic fusions) with the large CRV was also affected by the over-expression of this GTPase. Interestingly, cell overexpression of Rap2b wt markedly decreased the levels of the v-SNARE, Vamp7, suggesting that this down-regulation impairs the homotypic and heterotypic fusions events of the Coxiella vacuole.


Asunto(s)
Coxiella burnetii/fisiología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Fiebre Q/metabolismo , Vacuolas/microbiología , Proteínas de Unión al GTP rap/metabolismo , Animales , Células CHO , Chlorocebus aethiops , Cricetulus , AMP Cíclico/metabolismo , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Fusión de Membrana , Fagosomas/metabolismo , Fagosomas/microbiología , Fiebre Q/microbiología , Vacuolas/metabolismo , Células Vero
10.
PLoS One ; 14(1): e0209820, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30640917

RESUMEN

Microtubules (Mts) are dynamic cytoskeleton structures that play a key role in vesicular transport. The Mts-mediated transport depends on motor proteins named kinesins and the dynein/dynactin motor complex. The Rab7 adapter protein FYCO1 controls the anterograde transport of the endocytic compartments through the interaction with the kinesin KIF5. Rab7 and its partner RILP induce the recruitment of dynein/dynactin to late endosomes regulating its retrograde transport to the perinuclear area to fuse with lysosomes. The late endosomal-lysosomal fusion is regulated by the HOPS complex through its interaction with RILP and the GTPase Arl8. Coxiella burnetii (Cb), the causative agent of Q fever, is an obligate intracellular pathogen, which generates a large compartment with autophagolysosomal characteristics named Cb-containing vacuole (CCV). The CCV forms through homotypic fusion between small non-replicative CCVs (nrCCV) and through heterotypic fusion with other compartments, such as endosomes and lysosomes. In this work, we characterise the role of Mts, motor proteins, RILP/Rab7 and Arl8 on the CCV biogenesis. The formation of the CCV was affected when either the dynamics and/or the acetylation state of Mts were modified. Similarly, the overexpression of the dynactin subunit non-functional mutants p150Glued and RILP led to the formation of small nrCCVs. This phenomenon is not observed in cells overexpressing WT proteins, the motor KIF5 or its interacting protein FYCO1. The formation of the CCV was normal in infected cells that overexpressed Arl8 alone or together with hVps41 (a HOPS subunit) or in cells co-overexpressing hVps41 and RILP. The dominant negative mutant of Arl8 and the non-functional hVps41 inhibited the formation of the CCV. When the formation of CCV was affected, the bacterial multiplication diminished. Our results suggest that nrCCVs recruit the molecular machinery that regulate the Mts-dependent retrograde transport, Rab7/RILP and the dynein/dynactin system, as well as the tethering processes such as HOPS complex and Arl8 to finally originate the CCV where C. burnetii multiplies.


Asunto(s)
Coxiella burnetii/metabolismo , Dineínas/metabolismo , Microtúbulos/metabolismo , Citoesqueleto de Actina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Transporte Biológico , Chlorocebus aethiops , Coxiella burnetii/patogenicidad , Citoesqueleto/metabolismo , Complejo Dinactina/metabolismo , Endosomas/metabolismo , Células HeLa , Humanos , Lisosomas/metabolismo , Microtúbulos/fisiología , Transporte de Proteínas/fisiología , Fiebre Q/metabolismo , Vacuolas/metabolismo , Células Vero , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión a GTP rab7
11.
Infect Immun ; 86(4)2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29339460

RESUMEN

Coxiella burnetii is an intracellular pathogen that replicates in a lysosome-derived vacuole. A determinant necessary for C. burnetii virulence is the Dot/Icm type IVB secretion system (T4SS). The Dot/Icm system delivers more than 100 proteins, called type IV effectors (T4Es), across the vacuolar membrane into the host cell cytosol. Several T4Es have been shown to be important for vacuolar biogenesis. Here, transposon (Tn) insertion sequencing technology (INSeq) was used to identify C. burnetii Nine Mile phase II mutants in an arrayed library, which facilitated the identification and clonal isolation of mutants deficient in 70 different T4E proteins. These effector mutants were screened in HeLa cells for deficiencies in Coxiella-containing vacuole (CCV) biogenesis. This screen identified and validated seven new T4Es that were important for vacuole biogenesis. Loss-of-function mutations in cbu0414 (coxH1), cbu0513, cbu0978 (cem3), cbu1387 (cem6), cbu1524 (caeA), cbu1752, or cbu2028 resulted in a small-vacuole phenotype. These seven mutant strains produced small CCVs in all cells tested, which included macrophage-like cells. The cbu2028::Tn mutant, though unable to develop large CCVs, had intracellular replication rates similar to the rate of the parental strain of C. burnetii, whereas the other six effector mutants defective in CCV biogenesis displayed significant reductions in intracellular replication. Vacuoles created by the cbu0513::Tn mutant did not accumulate lipidated microtubule-associated protein 1A/1B light chain 3 (LC3-II), suggesting a failure in fusion of the CCV with autophagosomes. These seven T4E proteins add to the growing repertoire of C. burnetii factors that contribute to CCV biogenesis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Coxiella burnetii/fisiología , Fiebre Q/metabolismo , Fiebre Q/microbiología , Autofagosomas/metabolismo , Sistemas de Secreción Bacterianos , Coxiella burnetii/genética , Coxiella burnetii/patogenicidad , Elementos Transponibles de ADN , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Humanos , Lisosomas/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Mutación , Transporte de Proteínas , Vacuolas/metabolismo
12.
Infect Immun ; 86(4)2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29311244

RESUMEN

Our previous study demonstrated that neutrophils play an important role in host defense against Coxiella burnetii infection in mice. In this study, avirulent strain C. burnetii Nine Mile phase II (NMII) was used to examine if C. burnetii can modulate mouse bone marrow-derived neutrophil apoptosis. The results indicated that NMII can inhibit neutrophil apoptosis. Western blotting demonstrated that caspase-3 cleavage was decreased in NMII-infected neutrophils, while phosphorylated mitogen-activated protein kinase (MAPK) p38 and extracellular signal-regulated kinase 1 (Erk1) were increased. Additionally, p38, Erk1/2, phosphoinositide 3-kinase (PI3K), or NF-κB inhibitors reduced the ability of NMII to inhibit neutrophil apoptosis. These results suggest that NMII-mediated inhibition of neutrophil apoptosis depends on its ability to activate neutrophil MAPK pathways. Antiapoptotic protein myeloid cell leukemia-1 (Mcl-1) was significantly increased in NMII-infected neutrophils, and an Mcl-1 inhibitor significantly reduced the ability of NMII to inhibit neutrophil apoptosis. Mcl-1 protein stability was enhanced by phosphorylation at Thr-163 by Erk, and the protein levels were regulated by p38, Erk, PI3K, and NF-κB. Furthermore, the observation that a type IV secretion system (T4SS)-deficient dotA mutant showed a significantly reduced ability to inhibit neutrophil apoptosis compared to wild-type (WT) NMII suggests that T4SS-secreted factors may be involved in NMII-induced inhibition of neutrophil apoptosis. Collectively, these results demonstrate that NMII inhibits neutrophil apoptosis through inhibition of caspase-3 cleavage and activation of MAPK survival pathways with subsequent expression and stabilization of antiapoptotic protein Mcl-1, a process that may partially require the T4SS.


Asunto(s)
Apoptosis , Coxiella burnetii/inmunología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Neutrófilos/inmunología , Fiebre Q/inmunología , Fiebre Q/metabolismo , Transducción de Señal , Animales , Apoptosis/genética , Caspasa 3/metabolismo , Fragmentación del ADN , Modelos Animales de Enfermedad , Expresión Génica , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/microbiología , Ratones , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , FN-kappa B/metabolismo , Neutrófilos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteolisis , Fiebre Q/genética , Fiebre Q/microbiología , Sistemas de Secreción Tipo IV/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
PLoS One ; 12(3): e0173528, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28278296

RESUMEN

Published data show that murine bone marrow-derived macrophages (BMDM) restrict growth of avirulent phase II, but not virulent phase I, Coxiella burnetii. Growth restriction of phase II bacteria is thought to result from potentiated recognition of pathogen-associated molecular patterns, which leads to production of inhibitory effector molecules. Past studies have used conditioned medium from L-929 murine fibroblasts as a source of macrophage-colony stimulating factor (M-CSF) to promote differentiation of bone marrow-derived myeloid precursors into macrophages. However, uncharacterized components of conditioned medium, such as variable amounts of type I interferons, can affect macrophage activation status and their permissiveness for infection. In the current study, we show that the C. burnetii Nine Mile phase II (NMII) strain grows robustly in primary macrophages from C57BL/6J mice when bone marrow cells are differentiated with recombinant murine M-CSF (rmM-CSF). Bacteria were readily internalized by BMDM, and replicated within degradative, LAMP1-positive vacuoles to achieve roughly 3 logs of growth over 6 days. Uninfected BMDM did not appreciably express CD38 or Egr2, markers of classically (M1) and alternatively (M2) activated macrophages, respectively, nor did infection change the lack of polarization. In accordance with an M0 phenotype, infected BMDM produced moderate amounts of TNF and nitric oxide. Similar NMII growth results were obtained using C57BL/6J myeloid progenitors immortalized with an estrogen-regulated Hoxb8 (ER-Hoxb8) oncogene. To demonstrate the utility of the ER-Hoxb8 system, myeloid progenitors from natural resistance-associated macrophage protein 1 (Nramp1) C57BL/6J knock-in mice were transduced with ER-Hoxb8, and macrophages were derived from immortalized progenitors using rmM-CSF and infected with NMII. No difference in growth was observed when compared to macrophages from wild type mice, indicating depletion of metal ions by the Nramp1 transporter does not negatively impact NMII growth. Results with NMII were recapitulated in primary macrophages where C57BL/6J Nramp1+ BMDM efficiently killed Salmonella enterica serovar Typhimurium. M-CSF differentiated murine macrophages from bone marrow and conditional ER-Hoxb8 myeloid progenitors will be useful ex vivo models for studying Coxiella-macrophage interactions.


Asunto(s)
Médula Ósea/microbiología , Coxiella burnetii/crecimiento & desarrollo , Macrófagos/microbiología , Fiebre Q/microbiología , Animales , Médula Ósea/metabolismo , Células Cultivadas , Factores Estimulantes de Colonias/metabolismo , Coxiella burnetii/patogenicidad , Femenino , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Fiebre Q/metabolismo , Fiebre Q/patología , Factor de Necrosis Tumoral alfa/metabolismo
14.
PLoS Pathog ; 12(10): e1005915, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27711191

RESUMEN

Coxiella burnetii is an intracellular bacterial pathogen that causes human Q fever, an acute flu-like illness that can progress to chronic endocarditis and liver and bone infections. Humans are typically infected by aerosol-mediated transmission, and C. burnetii initially targets alveolar macrophages wherein the pathogen replicates in a phagolysosome-like niche known as the parasitophorous vacuole (PV). C. burnetii manipulates host cAMP-dependent protein kinase (PKA) signaling to promote PV formation, cell survival, and bacterial replication. In this study, we identified the actin regulatory protein vasodilator-stimulated phosphoprotein (VASP) as a PKA substrate that is increasingly phosphorylated at S157 and S239 during C. burnetii infection. Avirulent and virulent C. burnetii triggered increased levels of phosphorylated VASP in macrophage-like THP-1 cells and primary human alveolar macrophages, and this event required the Cα subunit of PKA. VASP phosphorylation also required bacterial protein synthesis and secretion of effector proteins via a type IV secretion system, indicating the pathogen actively triggers prolonged VASP phosphorylation. Optimal PV formation and intracellular bacterial replication required VASP activity, as siRNA-mediated depletion of VASP reduced PV size and bacterial growth. Interestingly, ectopic expression of a phospho-mimetic VASP (S239E) mutant protein prevented optimal PV formation, whereas VASP (S157E) mutant expression had no effect. VASP (S239E) expression also prevented trafficking of bead-containing phagosomes to the PV, indicating proper VASP activity is critical for heterotypic fusion events that control PV expansion in macrophages. Finally, expression of dominant negative VASP (S157A) in C. burnetii-infected cells impaired PV formation, confirming importance of the protein for proper infection. This study provides the first evidence of VASP manipulation by an intravacuolar bacterial pathogen via activation of PKA in human macrophages.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Interacciones Huésped-Patógeno/fisiología , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/microbiología , Proteínas de Microfilamentos/metabolismo , Fosfoproteínas/metabolismo , Fiebre Q/metabolismo , Coxiella burnetii , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Activación Enzimática/fisiología , Técnicas de Silenciamiento del Gen , Humanos , Immunoblotting , Inmunoprecipitación , Microscopía Confocal
15.
Infect Immun ; 84(9): 2524-33, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27324482

RESUMEN

Coxiella burnetii, the etiological agent of Q fever in humans, is an intracellular pathogen that replicates in an acidified parasitophorous vacuole derived from host lysosomes. Generation of this replicative compartment requires effectors delivered into the host cell by the Dot/Icm type IVb secretion system. Several effectors crucial for C. burnetii intracellular replication have been identified, but the host pathways coopted by these essential effectors are poorly defined, and very little is known about how spacious vacuoles are formed and maintained. Here we demonstrate that the essential type IVb effector, CirA, stimulates GTPase activity of RhoA. Overexpression of CirA in mammalian cells results in cell rounding and stress fiber disruption, a phenotype that is rescued by overexpression of wild-type or constitutively active RhoA. Unlike other effector proteins that subvert Rho GTPases to modulate uptake, CirA is the first effector identified that is dispensable for uptake and instead recruits Rho GTPase to promote biogenesis of the bacterial vacuole. Collectively our results highlight the importance of CirA in coopting host Rho GTPases for establishment of Coxiella burnetii infection and virulence in mammalian cell culture and mouse models of infection.


Asunto(s)
Proteínas Bacterianas/metabolismo , Coxiella burnetii/metabolismo , GTP Fosfohidrolasas/metabolismo , Fiebre Q/metabolismo , Sistemas de Secreción Tipo IV/metabolismo , Virulencia/fisiología , Proteína de Unión al GTP rhoA/metabolismo , Animales , Línea Celular Tumoral , Células HeLa , Interacciones Huésped-Patógeno/fisiología , Humanos , Lisosomas/metabolismo , Ratones , Transporte de Proteínas/fisiología , Fiebre Q/microbiología , Vacuolas/metabolismo , Vacuolas/microbiología
16.
Infect Immun ; 84(4): 940-949, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26787722

RESUMEN

Coxiella burnetii, the causative agent of Q fever, is an obligate intracellular, primarily pulmonary, bacterial pathogen. Although much is known about adaptive immune responses against this bacterium, our understanding of innate immune responses against C. burnetii is not well defined, particularly within the target tissue for infection, the lung. Previous studies examined the roles of the innate immune system receptors Toll-like receptor 2 (TLR2) and TLR4 in peripheral infection models and described minimal phenotypes in specific gene deletion animals compared to those of their wild-type controls (S. Meghari et al., Ann N Y Acad Sci 1063:161-166, 2005,http://dx.doi.org/10.1196/annals.1355.025; A. Honstettre et al., J Immunol 172:3695-3703, 2004,http://dx.doi.org/10.4049/jimmunol.172.6.3695) . Here, we assessed the roles for TLR2, TLR4, and MyD88 in pulmonary C. burnetii infection and compared responses to those that occurred in TLR2- and TLR4-deficient animals following peripheral infection. As observed previously, neither TLR2 nor TLR4 was needed for limiting bacterial growth after peripheral infection. In contrast, TLR2 and, to a lesser extent, TLR4 limited growth (or dissemination) of the bacterium in the lung and spleen after pulmonary infection. TLR2, TLR4, and MyD88 were not required for the general inflammatory response in the lungs after pulmonary infection. However, MyD88 signaling was important for infection-induced morbidity. Finally, TLR2 expression on hematopoietic cells was most important for limiting bacterial growth in the lung. These results expand on our knowledge of the roles for TLR2 and TLR4 in C. burnetii infection and suggest various roles for these receptors that are dictated by the site of infection.


Asunto(s)
Coxiella burnetii/fisiología , Enfermedades Pulmonares/microbiología , Factor 88 de Diferenciación Mieloide/metabolismo , Fiebre Q/metabolismo , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Quimera , Regulación de la Expresión Génica/inmunología , Regulación de la Expresión Génica/fisiología , Enfermedades Pulmonares/metabolismo , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Peritonitis/microbiología , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética
17.
PLoS One ; 10(4): e0124664, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25909586

RESUMEN

The obligate intracellular bacteria, Rickettsia rickettsii and Coxiella burnetii, are the potential agents of bio-warfare/bio-terrorism. Here C3H/HeN mice were immunized with a recombinant protein fragment rOmp-4 derived from outer membrane protein B, a major protective antigen of R. rickettsii, combined with chloroform-methanol residue (CMR) extracted from phase I C. burnetii organisms, a safer Q fever vaccine. These immunized mice had significantly higher levels of IgG1 and IgG2a to rOmpB-4 and interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α), two crucial cytokines in resisting intracellular bacterial infection, as well as significantly lower rickettsial loads and slighter pathological lesions in organs after challenge with R. rickettsii, compared with mice immunized with rOmpB-4 or CMR alone. Additionally, after challenge with C. burnetii, the coxiella loads in the organs of these mice were significantly lower than those of mice immunized with rOmpB-4 alone. Our results prove that CMR could markedly potentiate enhance the rOmpB-4-specific immunoprotection by promoting specific and non-specific immunoresponses and the immunization with the protective antigen of R. rickettsii combined with CMR of C. burnetii could confer effective protection against infection of R. rickettsii or C. burnetii.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Vacunas Bacterianas/inmunología , Coxiella burnetii/inmunología , Fiebre Q/prevención & control , Proteínas Recombinantes/inmunología , Rickettsia rickettsii/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/genética , Línea Celular , Cloroformo/farmacología , Coxiella burnetii/efectos de los fármacos , Citocinas/sangre , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Inmunización , Masculino , Metanol/farmacología , Ratones , Pruebas de Neutralización , Fiebre Q/inmunología , Fiebre Q/metabolismo , Fiebre Q/patología , Proteínas Recombinantes/genética
18.
Infect Immun ; 82(9): 3740-52, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24958706

RESUMEN

Coxiella burnetii, the causative agent of Q fever, is a human intracellular pathogen that utilizes the Icm/Dot type IVB secretion system to translocate effector proteins into host cells. To identify novel C. burnetii effectors, we applied a machine-learning approach to predict C. burnetii effectors, and examination of 20 such proteins resulted in the identification of 13 novel effectors. To determine whether these effectors, as well as several previously identified effectors, modulate conserved eukaryotic pathways, they were expressed in Saccharomyces cerevisiae. The effects on yeast growth were examined under regular growth conditions and in the presence of caffeine, a known modulator of the yeast cell wall integrity (CWI) mitogen-activated protein (MAP) kinase pathway. In the presence of caffeine, expression of the effectors CBU0885 and CBU1676 caused an enhanced inhibition of yeast growth, and the growth inhibition of CBU0388 was suppressed. Furthermore, analysis of synthetic lethality effects and examination of the activity of the CWI MAP kinase transcription factor Rlm1 indicated that CBU0388 enhances the activation of this MAP kinase pathway in yeast, while CBU0885 and CBU1676 abolish this activation. Additionally, coexpression of CBU1676 and CBU0388 resulted in mutual suppression of their inhibition of yeast growth. These results strongly indicate that these three effectors modulate the CWI MAP kinase pathway in yeast. Moreover, both CBU1676 and CBU0885 were found to contain a conserved haloacid dehalogenase (HAD) domain, which was found to be required for their activity. Collectively, our results demonstrate that MAP kinase pathways are most likely targeted by C. burnetii Icm/Dot effectors.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Coxiella burnetii/genética , Coxiella burnetii/metabolismo , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Transducción de Señal/genética , Línea Celular Tumoral , Células HL-60 , Humanos , Proteínas de Dominio MADS/genética , Proteínas de Dominio MADS/metabolismo , Transporte de Proteínas/genética , Fiebre Q/genética , Fiebre Q/metabolismo , Fiebre Q/microbiología , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
Artículo en Inglés | MEDLINE | ID: mdl-25566510

RESUMEN

The formation of granulomas is associated with the resolution of Q fever, a zoonosis due to Coxiella burnetii; however the molecular mechanisms of granuloma formation remain poorly understood. We generated human granulomas with peripheral blood mononuclear cells (PBMCs) and beads coated with C. burnetii, using BCG extracts as controls. A microarray analysis showed dramatic changes in gene expression in granuloma cells of which more than 50% were commonly modulated genes in response to C. burnetii and BCG. They included M1-related genes and genes related to chemotaxis. The inhibition of the chemokines, CCL2 and CCL5, directly interfered with granuloma formation. C. burnetii granulomas also expressed a specific transcriptional profile that was essentially enriched in genes associated with type I interferon response. Our results showed that granuloma formation is associated with a core of transcriptional response based on inflammatory genes. The specific granulomatous response to C. burnetii is characterized by the activation of type 1 interferon pathway.


Asunto(s)
Coxiella burnetii/fisiología , Granuloma/genética , Granuloma/microbiología , Fiebre Q/genética , Fiebre Q/microbiología , Adulto , Anciano , Coxiella burnetii/genética , Perfilación de la Expresión Génica , Granuloma/metabolismo , Humanos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/microbiología , Masculino , Persona de Mediana Edad , Fiebre Q/metabolismo
20.
Artículo en Inglés | MEDLINE | ID: mdl-25610812

RESUMEN

To survive in macrophages, Coxiella burnetii hijacks the activation pathway of macrophages. Recently, we have demonstrated that C. burnetii, via its lipopolysaccharide (LPS), avoids the activation of p38α-MAPK through an antagonistic engagement of Toll-like receptor (TLR)-4. We investigated the fine-tuned mechanism leading to the absence of activation of the p38α-MAPK despite TLR-4 engagement. In macrophages challenged with LPS from the avirulent variants of C. burnetii, TLR-4 and TLR-2 co-immunoprecipitated. This association was absent in cells challenged by the LPS of pathogenic C. burnetii. The disruption makes TLRs unable to signal during the recognition of the LPS of pathogenic C. burnetii. The disruption of TLR-2 and TLR-4 was induced by the re-organization of the macrophage cytoskeleton by C. burnetii LPS. Interestingly, blocking the actin cytoskeleton re-organization relieved the disruption of the association TLR-2/TLR-4 by pathogenic C. burnetii and rescued the p38α-MAPK activation by C. burnetii. We elucidated an unexpected mechanism allowing pathogenic C. burnetii to avoid macrophage activation by the disruption of the TLR-2 and TLR-4 association.


Asunto(s)
Coxiella burnetii/metabolismo , Lipopolisacáridos/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Fiebre Q/metabolismo , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Células Cultivadas , Coxiella burnetii/genética , Activación Enzimática , Interacciones Huésped-Patógeno , Humanos , Macrófagos/enzimología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 14 Activada por Mitógenos/genética , Unión Proteica , Fiebre Q/enzimología , Fiebre Q/genética , Fiebre Q/microbiología , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA