Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 152
Filtrar
1.
Virulence ; 15(1): 2350893, 2024 12.
Artigo em Inglês | MEDLINE | ID: mdl-38725096

RESUMO

Coxiella burnetii (C. burnetii) is the causative agent of Q fever, a zoonotic disease. Intracellular replication of C. burnetii requires the maturation of a phagolysosome-like compartment known as the replication permissive Coxiella-containing vacuole (CCV). Effector proteins secreted by the Dot/Icm secretion system are indispensable for maturation of a single large CCV by facilitating the fusion of promiscuous vesicles. However, the mechanisms of CCV maintenance and evasion of host cell clearance remain to be defined. Here, we show that C. burnetii secreted Coxiella vacuolar protein E (CvpE) contributes to CCV biogenesis by inducing lysosome-like vacuole (LLV) enlargement. LLV fission by tubulation and autolysosome degradation is impaired in CvpE-expressing cells. Subsequently, we found that CvpE suppresses lysosomal Ca2+ channel transient receptor potential channel mucolipin 1 (TRPML1) activity in an indirect manner, in which CvpE binds phosphatidylinositol 3-phosphate [PI(3)P] and perturbs PIKfyve activity in lysosomes. Finally, the agonist of TRPML1, ML-SA5, inhibits CCV biogenesis and C. burnetii replication. These results provide insight into the mechanisms of CCV maintenance by CvpE and suggest that the agonist of TRPML1 can be a novel potential treatment that does not rely on antibiotics for Q fever by enhancing Coxiella-containing vacuoles (CCVs) fission.


Assuntos
Proteínas de Bactérias , Coxiella burnetii , Lisossomos , Fosfatidilinositol 3-Quinases , Fosfatos de Fosfatidilinositol , Canais de Potencial de Receptor Transitório , Vacúolos , Coxiella burnetii/metabolismo , Coxiella burnetii/crescimento & desenvolvimento , Coxiella burnetii/genética , Vacúolos/microbiologia , Vacúolos/metabolismo , Lisossomos/metabolismo , Lisossomos/microbiologia , Fosfatos de Fosfatidilinositol/metabolismo , Humanos , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Canais de Potencial de Receptor Transitório/metabolismo , Canais de Potencial de Receptor Transitório/genética , Fosfatidilinositol 3-Quinases/metabolismo , Animais , Febre Q/microbiologia , Células HeLa , Interações Hospedeiro-Patógeno
2.
Adv Healthc Mater ; 13(7): e2302351, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38198823

RESUMO

Coxiella burnetti is an intracellular bacterium that causes Q fever, a disease of worldwide importance. Q-VAX® , the approved human Q fever vaccine, is a whole cell vaccine associated with safety concerns. Here a safe particulate subunit vaccine candidate is developed that is ambient-temperature stable and can be cost-effectively manufactured. Endotoxin-free Escherichia coli is bioengineered to efficiently self-assemble biopolymer particles (BPs) that are densely coated with either strings of 18 T-cell epitopes (COX-BP) or two full-length immunodominant antigens (YbgF-BP-Com1) all derived from C. burnetii. BP vaccine candidates are ambient-temperature stable. Safety and immunogenicity are confirmed in mice and guinea pig (GP) models. YbgF-BP-Com1 elicits specific and strong humoral immune responses in GPs with IgG titers that are at least 1 000 times higher than those induced by Q-VAX® . BP vaccine candidates are not reactogenic. After challenge with C. burnetii, YbgF-BP-Com1 vaccine leads to reduced fever responses and pathogen burden in the liver and the induction of proinflammatory cytokines IL-12 and IFN-γ inducible protein (IP-10) when compared to negative control groups. These data suggest that YbgF-BP-Com1 induces functional immune responses reducing infection by C. burnetii. Collectively, these findings illustrate the potential of BPs as effective antigen carrier for Q fever vaccine development.


Assuntos
Coxiella burnetii , Febre Q , Humanos , Animais , Camundongos , Cobaias , Febre Q/prevenção & controle , Coxiella burnetii/metabolismo , Vacinas Bacterianas , Imunidade , Vacinas de Subunidades Antigênicas/metabolismo
3.
Mol Microbiol ; 121(3): 513-528, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38115201

RESUMO

Intracellular bacteria have evolved mechanisms to invade host cells, establish an intracellular niche that allows survival and replication, produce progeny, and exit the host cell after completion of the replication cycle to infect new target cells. Bacteria exit their host cell by (i) initiation of apoptosis, (ii) lytic cell death, and (iii) exocytosis. While bacterial egress is essential for bacterial spreading and, thus, pathogenesis, we currently lack information about egress mechanisms for the obligate intracellular pathogen C. burnetii, the causative agent of the zoonosis Q fever. Here, we demonstrate that C. burnetii inhibits host cell apoptosis early during infection, but induces and/or increases apoptosis at later stages of infection. Only at later stages of infection did we observe C. burnetii egress, which depends on previously established large bacteria-filled vacuoles and a functional intrinsic apoptotic cascade. The released bacteria are not enclosed by a host cell membrane and can infect and replicate in new target cells. In summary, our data argue that C. burnetii egress in a non-synchronous way at late stages of infection. Apoptosis-induction is important for C. burnetii egress, but other pathways most likely contribute.


Assuntos
Coxiella burnetii , Febre Q , Humanos , Coxiella burnetii/metabolismo , Febre Q/metabolismo , Febre Q/microbiologia , Febre Q/patologia , Apoptose/fisiologia , Transdução de Sinais , Vacúolos/metabolismo , Interações Hospedeiro-Patógeno
4.
Arch Razi Inst ; 78(3): 785-796, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-38028822

RESUMO

Coxiella burnetii (C. burnetii), the etiological agent of the Q fever disease, ranks among the most sporadic and persistent global public health concerns. Ruminants are the principal source of human infections and diseases present in both acute and chronic forms. This bacterium is an intracellular pathogen that can survive and reproduce under acidic (pH 4 to 5) and harsh circumstances that contain Coxiella-containing vacuoles. By undermining the autophagy defense system of the host cell, C. burnetii is able to take advantage of the autophagy pathway, which allows it to improve the movement of nutrients and the membrane, thereby extending the vacuole of the reproducing bacteria. For this method to work, it requires the participation of many bacterial effector proteins. In addition, the precise and prompt identification of the causative agent of an acute disease has the potential to delay the onset of its chronic form. Moreover, to make accurate and rapid diagnoses, it is necessary to create diagnostic devices. This review summarizes the most recent research on the epidemiology, pathogenesis, and diagnosis approaches of C. burnetii. This study also explored the complicated relationships between C. burnetii and the autophagic pathway, which are essential for intracellular reproduction and survival in host cells for the infection to be effective.


Assuntos
Coxiella burnetii , Febre Q , Humanos , Coxiella burnetii/metabolismo , Febre Q/veterinária , Febre Q/metabolismo , Vacúolos/metabolismo , Vacúolos/microbiologia , Autofagia
5.
Int Rev Cell Mol Biol ; 377: 1-17, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37268347

RESUMO

In order to successfully establish a replicative niche, intracellular bacterial pathogens must influence eukaryotic cell biology. Vesicle and protein traffic, transcription and translation, metabolism and innate immune signaling are all important elements of the host-pathogen interaction that can be manipulated by intracellular bacterial pathogens. The causative agent of Q fever, Coxiella burnetii, is a mammalian adapted pathogen that replicates in a lysosome-derived pathogen-modified vacuole. C. burnetii establishes this replicative niche by using a cohort of novel proteins, termed effectors, to hijack the mammalian host cell. The functional and biochemical roles of a small number of effectors have been discovered and recent studies have demonstrated that mitochondria are a bona fide target for a subset of these effectors. Various approaches have begun to unravel the role these proteins play at mitochondria during infection, with key mitochondrial functions, including apoptosis and mitochondrial proteostasis, likely influenced by mitochondrially localized effectors. Additionally, mitochondrial proteins likely contribute to the host response to infection. Thus, investigating the interplay between host and pathogen elements at this central organelle will uncover important new understanding of the C. burnetii infection process. With the advent of new technologies and sophisticated omics approaches, we are poised to explore the interaction between host cell mitochondria and C. burnetii with unprecedented spatial and temporal resolution.


Assuntos
Coxiella burnetii , Febre Q , Animais , Humanos , Coxiella burnetii/metabolismo , Febre Q/metabolismo , Febre Q/microbiologia , Vacúolos/metabolismo , Vacúolos/microbiologia , Mitocôndrias/metabolismo , Interações Hospedeiro-Patógeno , Mamíferos
6.
Mol Microbiol ; 118(6): 744-764, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36385554

RESUMO

Coxiella burnetii is the causative agent of Q fever. All C. burnetii isolates encode either an autonomously replicating plasmid (QpH1, QpDG, QpRS, or QpDV) or QpRS-like chromosomally integrated plasmid sequences. The role of the ORFs present in these sequences is unknown. Here, the role of the ORFs encoded on QpH1 was investigated. Using a new C. burnetii shuttle vector (pB-TyrB-QpH1ori), we cured the C. burnetii Nine Mile Phase II strain of QpH1. The ΔQpH1 strain grew normally in axenic media but had a significant growth defect in Vero cells, indicating QpH1 was important for C. burnetii virulence. We developed an inducible CRISPR interference system to examine the role of individual QpH1 plasmid genes. CRISPRi of cbuA0027 resulted in significant growth defects in axenic media and THP-1 cells. The cbuA0028/cbuA0027 operon encodes CBUA0028 (ToxP) and CBUA0027 (AntitoxP), which are homologous to the HigB2 toxin and HigA2 antitoxin, respectively, from Vibrio cholerae. Consistent with toxin-antitoxin systems, overexpression of toxP resulted in a severe intracellular growth defect that was rescued by co-expression of antitoxP. ToxP inhibited protein translation. AntitoxP bound the toxP promoter (PtoxP) and ToxP, with the resulting complex binding also PtoxP. In summary, our data indicate that C. burnetii maintains an autonomously replicating plasmid because of a plasmid-based toxin-antitoxin system.


Assuntos
Coxiella burnetii , Sistemas Toxina-Antitoxina , Animais , Chlorocebus aethiops , Coxiella burnetii/genética , Coxiella burnetii/metabolismo , Sistemas Toxina-Antitoxina/genética , Células Vero , Plasmídeos/genética , Virulência
7.
Infect Immun ; 90(10): e0041022, 2022 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-36190257

RESUMO

Coxiella burnetii is an obligate intracellular bacterial pathogen that has evolved a unique biphasic developmental cycle. The infectious form of C. burnetii is the dormant small cell variant (SCV), which transitions to a metabolically active large cell variant (LCV) that replicates inside the lysosome-derived host vacuole. A Dot/Icm type IV secretion system (T4SS), which can deliver over 100 effector proteins to host cells, is essential for the biogenesis of the vacuole and intracellular replication. How the distinct C. burnetii life cycle impacts the assembly and function of the Dot/Icm T4SS has remained unknown. Here, we combine advanced cryo-focused ion beam (cryo-FIB) milling and cryo-electron tomography (cryo-ET) imaging to visualize all developmental transitions and the assembly of the Dot/Icm T4SS in situ. Importantly, assembled Dot/Icm machines were not present in the infectious SCV. The appearance of the assembled Dot/Icm machine correlated with the transition of the SCV to the LCV intracellularly. Furthermore, temporal characterization of C. burnetii morphological changes revealed regions of the inner membrane that invaginate to form tightly packed stacks during the LCV-to-SCV transition at late stages of infection, which may enable the SCV-to-LCV transition that occurs upon infection of a new host cell. Overall, these data establish how C. burnetii developmental transitions control critical bacterial processes to promote intracellular replication and transmission.


Assuntos
Coxiella burnetii , Coxiella burnetii/metabolismo , Sistemas de Secreção Tipo IV/metabolismo , Proteínas de Bactérias/metabolismo , Vacúolos/microbiologia , Lisossomos/metabolismo , Interações Hospedeiro-Patógeno
8.
Biol Cell ; 114(9): 237-253, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35851960

RESUMO

Ezrin protein is involved in the interaction of actin cytoskeleton with membrane receptors such as CD44. It regulates plasma membrane dynamics and intracellular signaling. Coxiella burnetii, the etiologic agent of Q fever, is internalized into host cell through a poorly characterized molecular mechanism. Here we analyzed the role of ezrin and CD44 in the C. burnetii internalization by HeLa cells. The knockdown of ezrin and CD44 inhibited the bacterial uptake. Interestingly, at early stages of C. burnetii internalization, ezrin was recruited to the cell membrane fraction and phosphorylated. Moreover, the overexpression of non-phosphorylatable and phosphomimetic ezrin mutants decreased and increased the bacterial entry, respectively. A decrease in the internalization of C. burnetii was observed by the overexpression of CD44 truncated forms containing the intracellular or the extracellular domains. Interestingly, the CD44 mutant was unable to interact with ERM proteins decreased the bacterial internalization. These findings demonstrate the participation of ezrin in the internalization process of C. burnetii in non-phagocytic cells. Additionally, we present evidence that CD44 receptor would be involved in that process.


Assuntos
Coxiella burnetii , Proteínas do Citoesqueleto/metabolismo , Receptores de Hialuronatos/metabolismo , Citoesqueleto de Actina , Coxiella burnetii/metabolismo , Células HeLa , Humanos
9.
Front Cell Infect Microbiol ; 12: 867689, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35755850

RESUMO

HIF1α is an important transcription factor regulating not only cellular responses to hypoxia, but also anti-infective defense responses. We recently showed that HIF1α hampers replication of the obligate intracellular pathogen Coxiella burnetii which causes the zoonotic disease Q fever. Prior to development of chronic Q fever, it is assumed that the bacteria enter a persistent state. As HIF1α and/or hypoxia might be involved in the induction of C. burnetii persistence, we analyzed the role of HIF1α and hypoxia in the interaction of macrophages with C. burnetii to understand how the bacteria manipulate HIF1α stability and activity. We demonstrate that a C. burnetii-infection initially induces HIF1α stabilization, which decreases then over the course of an infection. This reduction depends on bacterial viability and a functional type IV secretion system (T4SS). While neither the responsible T4SS effector protein(s) nor the molecular mechanism leading to this partial HIF1α destabilization have been identified, our results demonstrate that C. burnetii influences the expression of HIF1α target genes in multiple ways. Therefore, a C. burnetii infection promotes HIF1α-mediated upregulation of several metabolic target genes; affects apoptosis-regulators towards a more pro-apoptotic signature; and under hypoxic conditions, shifts the ratio of the inflammatory genes analyzed towards a pro-inflammatory profile. Taken together, C. burnetii modulates HIF1α in a still elusive manner and alters the expression of multiple HIF1α target genes.


Assuntos
Coxiella burnetii , Febre Q , Coxiella burnetii/metabolismo , Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Hipóxia , Febre Q/microbiologia , Sistemas de Secreção Tipo IV/metabolismo
10.
Anal Chem ; 94(12): 4988-4996, 2022 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-35302749

RESUMO

The life cycle of intracellular pathogens is often complex and can include different morphoforms. Treatment of intracellular infections and unperturbed studying of the pathogen inside the host cell are frequently challenging. Here, we present a Raman-based, label-free, non-invasive, and non-destructive method to localize, visualize, and even quantify intracellular bacteria in 3D within intact host cells in a Coxiella burnetii infection model. C. burnetii is a zoonotic obligate intracellular pathogen that causes infections in ruminant livestock and humans with an acute disease known as Q fever. Using statistical data analysis, no isolation is necessary to gain detailed information on the intracellular pathogen's metabolic state. High-quality false color image stacks with diffraction-limited spatial resolution enable a 3D spatially resolved single host cell analysis that shows excellent agreement with results from transmission electron microscopy. Quantitative analysis at different time points post infection allows to follow the infection cycle with the transition from the large cell variant (LCV) to the small cell variant (SCV) at around day 6 and a gradual change in the lipid composition during vacuole maturation. Spectral characteristics of intracellular LCV and SCV reveal a higher lipid content of the metabolically active LCV.


Assuntos
Coxiella burnetii , Coxiella burnetii/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Vacúolos
11.
PLoS Pathog ; 18(2): e1010266, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35134097

RESUMO

Inhibition of host cell apoptosis is crucial for survival and replication of several intracellular bacterial pathogens. To interfere with apoptotic pathways, some pathogens use specialized secretion systems to inject bacterial effector proteins into the host cell cytosol. One of these pathogens is the obligate intracellular bacterium Coxiella burnetii, the etiological agent of the zoonotic disease Q fever. In this study, we analyzed the molecular activity of the anti-apoptotic T4SS effector protein AnkG (CBU0781) to understand how C. burnetii manipulates host cell viability. We demonstrate by co- and RNA-immunoprecipitation that AnkG binds to the host cell DExD box RNA helicase 21 (DDX21) as well as to the host cell 7SK small nuclear ribonucleoprotein (7SK snRNP) complex, an important regulator of the positive transcription elongation factor b (P-TEFb). The co-immunoprecipitation of AnkG with DDX21 is probably mediated by salt bridges and is independent of AnkG-7SK snRNP binding, and vice versa. It is known that DDX21 facilitates the release of P-TEFb from the 7SK snRNP complex. Consistent with the documented function of released P-TEFb in RNA Pol II pause release, RNA sequencing experiments confirmed AnkG-mediated transcriptional reprogramming and showed that expression of genes involved in apoptosis, trafficking, and transcription are influenced by AnkG. Importantly, DDX21 and P-TEFb are both essential for AnkG-mediated inhibition of host cell apoptosis, emphasizing the significance of the interaction of AnkG with both, the DDX21 protein and the 7SK RNA. In line with a critical function of AnkG in pathogenesis, the AnkG deletion C. burnetii strain was severely affected in its ability to inhibit host cell apoptosis and to generate a replicative C. burnetii-containing vacuole. In conclusion, the interference with the activity of regulatory host cell RNAs mediated by a bacterial effector protein represent a novel mechanism through which C. burnetii modulates host cell transcription, thereby enhancing permissiveness to bacterial infection.


Assuntos
Proteínas de Bactérias/metabolismo , Coxiella burnetii/metabolismo , RNA Helicases DEAD-box/metabolismo , Fator B de Elongação Transcricional Positiva/metabolismo , Febre Q/metabolismo , Ribonucleoproteínas Nucleares Pequenas/metabolismo , Sistemas de Secreção Tipo IV/metabolismo , Apoptose , Sobrevivência Celular , Coxiella burnetii/genética , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Mutação , Febre Q/microbiologia , Células THP-1
12.
Mol Microbiol ; 117(2): 235-251, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34874584

RESUMO

Anti-bacterial autophagy, known as xenophagy, is a host innate immune response that targets invading pathogens for degradation. Some intracellular bacteria, such as the enteric pathogen Salmonella enterica serovar Typhimurium (S. Typhimurium), utilize effector proteins to interfere with autophagy. One such S. Typhimurium effector, SopF, inhibits recruitment of ATG16L1 to damaged Salmonella-containing vacuoles (SCVs), thereby inhibiting the host xenophagic response. SopF is also required to maintain the integrity of the SCV during the early stages of infection. Here we show disruption of the SopF-ATG16L1 interaction leads to an increased proportion of cytosolic S. Typhimurium. Furthermore, SopF was utilized as a molecular tool to examine the requirement for ATG16L1 in the intracellular lifestyle of Coxiella burnetii, a bacterium that requires a functional autophagy pathway to replicate efficiently and form a single, spacious vacuole called the Coxiella-containing vacuole (CCV). ATG16L1 is required for CCV expansion and fusion but does not influence C. burnetii replication. In contrast, SopF did not affect CCV formation or replication, demonstrating that the contribution of ATG16L1 to CCV biogenesis is via its role in autophagy, not xenophagy. This study highlights the diverse capabilities of bacterial effector proteins to dissect the molecular details of host-pathogen interactions.


Assuntos
Coxiella burnetii , Vacúolos , Proteínas Relacionadas à Autofagia/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Coxiella/metabolismo , Coxiella burnetii/metabolismo , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo , Vacúolos/metabolismo
13.
Pathog Dis ; 79(7)2021 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-34323972

RESUMO

We hypothesize that intracellular trafficking pathways are altered in chlamydial infected cells to maximize the ability of Chlamydia to scavenge nutrients while not overtly stressing the host cell. Previous data demonstrated the importance of two eukaryotic SNARE proteins, VAMP4 and syntaxin 10 (Stx10), in chlamydial growth and development. Although, the mechanism for these effects is still unknown. To interrogate whether chlamydial infection altered these proteins' networks, we created BirA*-VAMP4 and BirA*-Stx10 fusion constructs to use the BioID proximity labeling system. While we identified a novel eukaryotic protein-protein interaction between Stx10 and VAPB, we also identified caveats in using the BioID system to study the impact of infection by an obligate intracellular pathogen on SNARE protein networks. The addition of the BirA* altered the localization of VAMP4 and Stx10 during infection with Chlamydia trachomatis serovars L2 and D and Coxiella burnetii Nine Mile Phase II. We also discovered that BirA* traffics to and biotinylates Coxiella-containing vacuoles and, in general, has a propensity for labeling membrane or membrane-associated proteins. While the BioID system identified a novel association for Stx10, it is not a reliable methodology to examine intracellular trafficking pathway dynamics during infection with intracellular pathogens.


Assuntos
Infecções por Chlamydia/metabolismo , Chlamydia trachomatis/metabolismo , Coxiella burnetii/metabolismo , Proteoma/metabolismo , Proteínas Qa-SNARE/metabolismo , Proteínas R-SNARE/metabolismo , Proteínas SNARE/metabolismo , Proteínas de Bactérias/metabolismo , Biotinilação , Carbono-Nitrogênio Ligases/metabolismo , Proteínas de Escherichia coli/metabolismo , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Corpos de Inclusão/metabolismo , Mapeamento de Interação de Proteínas/métodos , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/metabolismo , Coloração e Rotulagem , Vacúolos/metabolismo
14.
PLoS One ; 16(4): e0249354, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33793664

RESUMO

Q fever is one of the most important zoonotic diseases caused by the obligate intracellular bacteria, Coxiella burnetii. This bacterial infection has been frequently reported in both humans and animals, especially ruminants. Ticks are important ectoparasite and serve as reservoir hosts of Coxiella-like endosymbionts (CLEs). In this study, we have attempted to express chaperone-coding genes from CLEs of Rhipicephalus annulatus ticks collected fromcow path. The partial DnaK coding sequence has been amplified and expressed by Escherichia coli. Amino acid sequences have been analyzed by MS-MS spectrometry and the UniProt database. Despites nucleotide sequences indicating high nucleotide variation and diversity, many nucleotide substitutions are synonymous. In addition, amino acid substitutions compensate for the physicochemical properties of the original amino acids. Immune Epitope Database and Analysis Resource (IEDB-AR) was employed to indicate the antigenicity of the partial DnaK protein and predict the epitopes of B-and T-cells. Interestingly, some predicted HLA-A and B alleles of the MHC-I and HLA-DR alleles belonging to MHC-II were similar to T-cell responses to C. burnetii in Q fever patients. Therefore, the partial DnaK protein of CLE from R. annulatus could be considered a vaccine candidate and immunogenic marker with future prospects.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Bactérias/metabolismo , Coxiella burnetii/metabolismo , Rhipicephalus/microbiologia , Adenosina Trifosfatases/classificação , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/imunologia , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/classificação , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Coxiella burnetii/isolamento & purificação , DNA Bacteriano/química , DNA Bacteriano/metabolismo , Bases de Dados Genéticas , Epitopos/análise , Epitopos/imunologia , Haplótipos , Mutação , Filogenia , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Simbiose
15.
Autophagy ; 17(3): 706-722, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32116095

RESUMO

Coxiella burnetii, the etiological agent of the zoonosis Q fever, replicates inside host cells within a large vacuole displaying autolysosomal characteristics. The development of this compartment is mediated by bacterial effectors, which interfere with a number of host membrane trafficking pathways. By screening a Coxiella transposon mutant library, we observed that transposon insertions in cbu0626 led to intracellular replication and vacuole biogenesis defects. Here, we demonstrate that CBU0626 is a novel member of the Coxiella vacuolar protein (Cvp) family of effector proteins, which is translocated by the Dot/Icm secretion system and localizes to vesicles with autolysosomal features as well as Coxiella-containing vacuoles (CCVs). We thus renamed this effector CvpF for Coxiella vacuolar protein F. CvpF specifically interacts with the host small GTPase RAB26, leading to the recruitment of the autophagosomal marker MAP1LC3B/LC3B (microtubule associated protein 1 light chain 3 beta) to CCVs. Importantly, cvpF::Tn mutants were highly attenuated compared to wild-type bacteria in the SCID mouse model of infection, highlighting the importance of CvpF for Coxiella virulence. These results suggest that CvpF manipulates endosomal trafficking and macroautophagy/autophagy induction for optimal C. burnetii vacuole biogenesis.Abbreviations: ACCM: acidified citrate cystein medium; AP: adaptor related protein complex; CCV: Coxiella-containing vacuole; Cvp: Coxiella vacuolar protein; GDI: guanosine nucleotide dissociation inhibitor; GDF: GDI dissociation factor; GEF: guanine exchange factor; LAMP1: lysosomal associated membrane protein 1; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MTORC1: mechanistic target of rapamycin kinase MTOR complex 1; PBS: phosphate-buffered saline; PMA: phorbol myristate acetate; SQSTM1/p62: sequestosome 1; WT: wild-type.


Assuntos
Autofagia/fisiologia , Sistemas de Secreção Bacterianos/metabolismo , Coxiella/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Vacúolos/microbiologia , Animais , Proteínas de Bactérias/metabolismo , Coxiella burnetii/crescimento & desenvolvimento , Coxiella burnetii/metabolismo , Humanos , Camundongos , Vacúolos/metabolismo
16.
Nat Microbiol ; 6(1): 19-26, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33139883

RESUMO

Gram-negative bacteria have a cell envelope that comprises an outer membrane (OM), a peptidoglycan (PG) layer and an inner membrane (IM)1. The OM and PG are load-bearing, selectively permeable structures that are stabilized by cooperative interactions between IM and OM proteins2,3. In Escherichia coli, Braun's lipoprotein (Lpp) forms the only covalent tether between the OM and PG and is crucial for cell envelope stability4; however, most other Gram-negative bacteria lack Lpp so it has been assumed that alternative mechanisms of OM stabilization are present5. We used a glycoproteomic analysis of PG to show that ß-barrel OM proteins are covalently attached to PG in several Gram-negative species, including Coxiella burnetii, Agrobacterium tumefaciens and Legionella pneumophila. In C. burnetii, we found that four different types of covalent attachments occur between OM proteins and PG, with tethering of the ß-barrel OM protein BbpA becoming most abundant in the stationary phase and tethering of the lipoprotein LimB similar throughout the cell cycle. Using a genetic approach, we demonstrate that the cell cycle-dependent tethering of BbpA is partly dependent on a developmentally regulated L,D-transpeptidase (Ldt). We use our findings to propose a model of Gram-negative cell envelope stabilization that includes cell cycle control and an expanded role for Ldts in covalently attaching surface proteins to PG.


Assuntos
Agrobacterium tumefaciens/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Coxiella burnetii/metabolismo , Escherichia coli/metabolismo , Legionella pneumophila/metabolismo , Peptidoglicano/metabolismo , Ciclo Celular/fisiologia , Membrana Celular/metabolismo , Parede Celular/metabolismo , Lipoproteínas/metabolismo , Simulação de Dinâmica Molecular , Peptidil Transferases/metabolismo , Ligação Proteica/fisiologia
17.
Sci Rep ; 10(1): 15396, 2020 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-32958854

RESUMO

The ability to inhibit host cell apoptosis is important for the intracellular replication of the obligate intracellular pathogen Coxiella burnetii, as it allows the completion of the lengthy bacterial replication cycle. Effector proteins injected into the host cell by the C. burnetii type IVB secretion system (T4BSS) are required for the inhibition of host cell apoptosis. AnkG is one of these anti-apoptotic effector proteins. The inhibitory effect of AnkG requires its nuclear localization, which depends on p32-dependent intracellular trafficking and importin-α1-mediated nuclear entry of AnkG. Here, we compared the sequences of ankG from 37 C. burnetii isolates and classified them in three groups based on the predicted protein size. The comparison of the three different groups allowed us to identify the first 28 amino acids as essential and sufficient for the anti-apoptotic activity of AnkG. Importantly, only the full-length protein from the first group is a bona fide effector protein injected into host cells during infection and has anti-apoptotic activity. Finally, using the Galleria mellonella infection model, we observed that AnkG from the first group has the ability to attenuate pathology during in vivo infection, as it allows survival of the larvae despite bacterial replication.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Proteínas de Bactérias/fisiologia , Morte Celular/efeitos dos fármacos , Coxiella burnetii/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Transporte Proteico , Alinhamento de Sequência , Fatores de Virulência/metabolismo
18.
Cell Microbiol ; 22(10): e13246, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32720355

RESUMO

Intracellular bacterial pathogens harbour genes, the closest homologues of which are found in eukaryotes. Regulator of chromosome condensation 1 (RCC1) repeat proteins are phylogenetically widespread and implicated in protein-protein interactions, such as the activation of the small GTPase Ran by its cognate guanine nucleotide exchange factor, RCC1. Legionella pneumophila and Coxiella burnetii, the causative agents of Legionnaires' disease and Q fever, respectively, harbour RCC1 repeat coding genes. Legionella pneumophila secretes the RCC1 repeat 'effector' proteins LegG1, PpgA and PieG into eukaryotic host cells, where they promote the activation of the pleiotropic small GTPase Ran, microtubule stabilisation, pathogen vacuole motility and intracellular bacterial growth as well as host cell migration. The RCC1 repeat effectors localise to the pathogen vacuole or the host plasma membrane and target distinct components of the Ran GTPase cycle, including Ran modulators and the small GTPase itself. Coxiella burnetii translocates the RCC1 repeat effector NopA into host cells, where the protein localises to nucleoli. NopA binds to Ran GTPase and promotes the nuclear accumulation of Ran(GTP), thus pertubing the import of the transcription factor NF-κB and innate immune signalling. Hence, divergent evolution of bacterial RCC1 repeat effectors defines the range of Ran GTPase cycle targets and likely allows fine-tuning of Ran GTPase activation by the pathogens at different cellular sites.


Assuntos
Evolução Biológica , Coxiella burnetii/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Legionella pneumophila/metabolismo , Proteína ran de Ligação ao GTP/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Nucléolo Celular/enzimologia , Coxiella burnetii/genética , Coxiella burnetii/patogenicidade , Ativação Enzimática , Genes Bacterianos , Interações Hospedeiro-Patógeno , Humanos , Legionella/genética , Legionella/metabolismo , Legionella/patogenicidade , Legionella pneumophila/genética , Doença dos Legionários/microbiologia , Transporte Proteico , Febre Q/microbiologia , Vacúolos/metabolismo , Vacúolos/microbiologia
19.
Proc Natl Acad Sci U S A ; 117(24): 13708-13718, 2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32482853

RESUMO

The Q fever agent Coxiella burnetii uses a defect in organelle trafficking/intracellular multiplication (Dot/Icm) type 4b secretion system (T4SS) to silence the host innate immune response during infection. By investigating C. burnetii effector proteins containing eukaryotic-like domains, here we identify NopA (nucleolar protein A), which displays four regulator of chromosome condensation (RCC) repeats, homologous to those found in the eukaryotic Ras-related nuclear protein (Ran) guanine nucleotide exchange factor (GEF) RCC1. Accordingly, NopA is found associated with the chromatin nuclear fraction of cells and uses the RCC-like domain to interact with Ran. Interestingly, NopA triggers an accumulation of Ran-GTP, which accumulates at nucleoli of transfected or infected cells, thus perturbing the nuclear import of transcription factors of the innate immune signaling pathway. Accordingly, qRT-PCR analysis on a panel of cytokines shows that cells exposed to the C. burnetii nopA::Tn or a Dot/Icm-defective dotA::Tn mutant strain present a functional innate immune response, as opposed to cells exposed to wild-type C. burnetii or the corresponding nopA complemented strain. Thus, NopA is an important regulator of the innate immune response allowing Coxiella to behave as a stealth pathogen.


Assuntos
Proteínas de Bactérias/metabolismo , Coxiella burnetii/metabolismo , Febre Q/imunologia , Animais , Proteínas de Bactérias/genética , Coxiella burnetii/genética , Feminino , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Febre Q/genética , Febre Q/microbiologia
20.
Cell Microbiol ; 22(5): e13154, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31872956

RESUMO

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.


Assuntos
Coxiella burnetii/metabolismo , NADP/metabolismo , Estresse Oxidativo , Coxiella burnetii/crescimento & desenvolvimento , Citoplasma/metabolismo , Células HeLa , Humanos , Macrófagos/microbiologia , Mutação , NADP/genética , Febre Q/metabolismo , Febre Q/microbiologia , Regeneração , Vacúolos/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA