Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Commun Biol ; 6(1): 1010, 2023 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-37798443

RESUMO

Pyroptosis is a cell death process that causes inflammation and contributes to numerous diseases. Pyroptosis is mediated by caspase-1 family proteases that cleave the pore-forming protein gasdermin D, causing plasma membrane rupture and release of pathogenic cellular contents. We previously identified muscimol as a small molecule that prevents plasma membrane rupture during pyroptosis via an unidentified mechanism. Here, we show that muscimol has reversible activity to prevent cellular lysis without affecting earlier pyroptotic events. Although muscimol is a well-characterized agonist for neuronal GABAA receptors, muscimol protection is not altered by GABAA receptor antagonists or recapitulated by other GABAA agonists, suggesting that muscimol acts via a novel mechanism. We find that muscimol blocks oligomerization of ninjurin-1, which is required for plasma membrane rupture downstream of gasdermin D pore formation. Our structure-activity relationship studies reveal distinct molecular determinants defining inhibition of pyroptotic lysis compared to GABAA binding. In addition, we demonstrate that muscimol reduces lethality during LPS-induced septic shock. Together, these findings demonstrate that ninjurin-1-mediated plasma membrane rupture can be pharmacologically modulated and pave the way toward identification of therapeutic strategies for pathologic conditions associated with pyroptosis.


Assuntos
Gasderminas , Piroptose , Muscimol/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Membrana Celular/metabolismo , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Ácido gama-Aminobutírico/metabolismo
2.
Methods Mol Biol ; 2641: 1-16, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37074637

RESUMO

Pyroptosis is a regulated form of cell death that leads to inflammation and plays a role in many different diseases. Pyroptosis was initially defined by the dependence on caspase-1, a protease which is activated by innate immune signaling complexes called inflammasomes. Caspase-1 cleaves the protein gasdermin D, releasing the N-terminal pore-forming domain, which inserts into the plasma membrane. Recent studies have revealed that other gasdermin family members form plasma membrane pores, leading to lytic cell death, and the definition of pyroptosis was revised to gasdermin-dependent cell death. In this review, we discuss how the use of the term pyroptosis has changed over time, as well as currently understood molecular mechanisms leading to pyroptosis and functional consequences of this form of regulated cell death.


Assuntos
Gasderminas , Piroptose , Inflamassomos/metabolismo , Morte Celular , Caspase 1/metabolismo
3.
Methods Mol Biol ; 2543: 179-189, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36087268

RESUMO

Pyroptosis is a highly regulated inflammatory form of cell death that plays a role in many different diseases, including cancer. Pyroptosis was initially described to be mediated by caspase-1, which is activated by innate immune signaling complexes called inflammasomes. Inflammasomes trigger caspase-dependent activation of the pore-forming protein, gasdermin D, and plasma membrane disruption. In this protocol, we describe a method to simultaneously detect two hallmarks of inflammasome-mediated pyroptosis. Using a fluorescently tagged inflammasome adaptor protein (ASC-Citrine) and membrane-impermeable nuclear dyes, we can track inflammasome formation and plasma membrane disruption over time in the same cell population.


Assuntos
Inflamassomos , Piroptose , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Caspase 1/metabolismo , Inflamassomos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo
4.
Cell Death Dis ; 10(4): 326, 2019 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-30975978

RESUMO

Pyroptosis is a programmed process of proinflammatory cell death mediated by caspase-1-related proteases that cleave the pore-forming protein, gasdermin D, causing cell lysis and release of inflammatory intracellular contents. The amino acid glycine prevents pyroptotic lysis via unknown mechanisms, without affecting caspase-1 activation or pore formation. Pyroptosis plays a critical role in diverse inflammatory diseases, including sepsis. Septic lethality is prevented by glycine treatment, suggesting that glycine-mediated cytoprotection may provide therapeutic benefit. In this study, we systematically examined a panel of small molecules, structurally related to glycine, for their ability to prevent pyroptotic lysis. We found a requirement for the carboxyl group, and limited tolerance for larger amino groups and substitution of the hydrogen R group. Glycine is an agonist for the neuronal glycine receptor, which acts as a ligand-gated chloride channel. The array of cytoprotective small molecules we identified resembles that of known glycine receptor modulators. However, using genetically deficient Glrb mutant macrophages, we found that the glycine receptor is not required for pyroptotic cytoprotection. Furthermore, protection against pyroptotic lysis is independent of extracellular chloride conductance, arguing against an effect mediated by ligand-gated chloride channels. Finally, we conducted a small-scale, hypothesis-driven small-molecule screen and identified unexpected ion channel modulators that prevent pyroptotic lysis with increased potency compared to glycine. Together, these findings demonstrate that pyroptotic lysis can be pharmacologically modulated and pave the way toward identification of therapeutic strategies for pathologic conditions associated with pyroptosis.


Assuntos
Citoproteção/efeitos dos fármacos , Glicina/análogos & derivados , Glicina/química , Macrófagos/efeitos dos fármacos , Piroptose/fisiologia , Animais , Antígenos de Bactérias/toxicidade , Toxinas Bacterianas/toxicidade , Caspase 1/metabolismo , Morte Celular , Células Cultivadas , Glicina/metabolismo , Canais Iônicos/metabolismo , Canais Iônicos/fisiologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Receptores de Glicina/agonistas , Receptores de Glicina/antagonistas & inibidores , Receptores de Glicina/metabolismo , Salmonella
5.
Proc Natl Acad Sci U S A ; 116(11): 5061-5070, 2019 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-30796192

RESUMO

Pyroptosis is an inflammatory form of programmed cell death following cellular damage or infection. It is a lytic process driven by gasdermin D-mediated cellular permeabilization and presumed osmotic forces thought to induce swelling and rupture. We found that pyroptotic cells do not spontaneously rupture in culture but lose mechanical resilience. As a result, cells were susceptible to rupture by extrinsic forces, such as shear stress or compression. Cell analyses revealed that all major cytoskeleton components were disrupted during pyroptosis and that sensitivity to rupture was calpain-dependent and linked with cleavage of vimentin and loss of intermediate filaments. Moreover, while release of lactate dehydrogenase (LDH), HMGB1, and IL-1ß occurred without rupture, rupture was required for release of large inflammatory stimuli-ASC specks, mitochondria, nuclei, and bacteria. Importantly, supernatants from ruptured cells were more immunostimulatory than those from nonruptured cells. These observations reveal undiscovered cellular events occurring during pyroptosis, define the mechanisms driving pyroptotic rupture, and highlight the immunologic importance of this event.


Assuntos
Calpaína/metabolismo , Imunização , Filamentos Intermediários/metabolismo , Piroptose , Vimentina/metabolismo , Alarminas/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Caspase 1/metabolismo , Força Compressiva , Citoesqueleto/metabolismo , Citosol/metabolismo , Humanos , Inflamassomos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Neoplasias/metabolismo , Proteínas de Ligação a Fosfato , Estresse Mecânico , Células THP-1
6.
Curr Protoc Immunol ; 122(1): e52, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30028908

RESUMO

Pyroptosis is a form of programmed pro-inflammatory cell death that plays a protective role in the host response to infection, but can also promote pathogenic inflammation. Pyroptosis is mediated by the cysteine protease, caspase-1. Caspase-1 cleaves gasdermin D, releasing the N-terminal pore-forming domain, which inserts into the plasma membrane and drives osmotic lysis. Caspase-1 also proteolytically activates the inflammatory cytokines interleukin 1ß (IL-1ß) and IL-18. This unit describes methods for stimulating pyroptosis and assessing subsequent loss of plasma membrane integrity. We also describe an ELISA to quantify released IL-1ß. These methods can be applied to many different types of experiments. © 2018 by John Wiley & Sons, Inc.

7.
J Vis Exp ; (135)2018 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-29863661

RESUMO

Inflammasomes are innate immune signaling platforms that are required for the successful control of many pathogenic organisms, but also promote inflammatory and autoinflammatory diseases. Inflammasomes are activated by cytosolic pattern recognition receptors, including members of the NOD-like receptor (NLR) family. These receptors oligomerize upon the detection of microbial or damage-associated stimuli. Subsequent recruitment of the adaptor protein ASC forms a microscopically visible inflammasome complex, which activates caspase-1 through proximity-induced auto-activation. Following the activation, caspase-1 cleaves pro-IL-1ß and pro-IL-18, leading to the activation and secretion of these pro-inflammatory cytokines. Caspase-1 also mediates the inflammatory form of cell death termed pyroptosis, which features the loss of membrane integrity and cell lysis. Caspase-1 cleaves gasdermin D, releasing the N-terminal fragment which forms plasma membrane pores, leading to osmotic lysis. In vitro, the activation of caspase-1 can be determined by labeling bone marrow-derived macrophages with the caspase-1 activity probe FAM-YVAD-FMK and by labeling the cells with antibodies against the adaptor protein ASC. This technique allows the identification of inflammasome formation and caspase-1 activation in individual cells using fluorescence microscopy. Pyroptotic cell death can be detected by measuring the release of cytosolic lactate dehydrogenase into the medium. This procedure is simple, cost effective and performed in a 96-well plate format, allowing adaptation for screening. In this manuscript, we show that activation of the NLRP3 inflammasome by nigericin leads to the co-localization of the adaptor protein ASC and active caspase-1, leading to pyroptosis.


Assuntos
Inflamassomos/imunologia , Macrófagos/metabolismo , Piroptose/genética , Animais , Imunidade Inata , Camundongos , Transdução de Sinais
8.
Cell Host Microbe ; 14(2): 159-70, 2013 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-23954155

RESUMO

Eradication of persistent intracellular bacterial pathogens with antibiotic therapy is often slow or incomplete. However, strategies to augment antibiotics are hampered by our poor understanding of the nutritional environment that sustains chronic infection. Here we show that the intracellular pathogen Brucella abortus survives and replicates preferentially in alternatively activated macrophages (AAMs), which are more abundant during chronic infection. A metabolic shift induced by peroxisome proliferator-activated receptor γ (PPARγ), which increases intracellular glucose availability, is identified as a causal mechanism promoting enhanced bacterial survival in AAMs. Glucose uptake was crucial for increased replication of B. abortus in AAMs, and for chronic infection, as inactivation of the bacterial glucose transporter gluP reduced both intracellular survival in AAMs and persistence in mice. Thus, a shift in intracellular nutrient availability induced by PPARγ promotes chronic persistence of B. abortus within AAMs, and targeting this pathway may aid in eradicating chronic infection.


Assuntos
Brucella abortus/fisiologia , Glucose/metabolismo , Ativação de Macrófagos , Macrófagos/microbiologia , Viabilidade Microbiana , PPAR gama/metabolismo , Animais , Brucella abortus/crescimento & desenvolvimento , Brucella abortus/imunologia , Brucella abortus/metabolismo , Macrófagos/imunologia , Camundongos
9.
mBio ; 4(1): e00418-12, 2013 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-23422410

RESUMO

Host cytokine responses to Brucella abortus infection are elicited predominantly by the deployment of a type IV secretion system (T4SS). However, the mechanism by which the T4SS elicits inflammation remains unknown. Here we show that translocation of the T4SS substrate VceC into host cells induces proinflammatory responses. Ectopically expressed VceC interacted with the endoplasmic reticulum (ER) chaperone BiP/Grp78 and localized to the ER of HeLa cells. ER localization of VceC required a transmembrane domain in its N terminus. Notably, the expression of VceC resulted in reorganization of ER structures. In macrophages, VceC was required for B. abortus-induced inflammation by induction of the unfolded protein response by a process requiring inositol-requiring transmembrane kinase/endonuclease 1. Altogether, these findings suggest that translocation of the T4SS effector VceC induces ER stress, which results in the induction of proinflammatory host cell responses during B. abortus infection. IMPORTANCE Brucella species are pathogens that require a type IV secretion system (T4SS) to survive in host cells and to maintain chronic infection. By as-yet-unknown pathways, the T4SS also elicits inflammatory responses in infected cells. Here we show that inflammation caused by the T4SS results in part from the sensing of a T4SS substrate, VceC, that localizes to the endoplasmic reticulum (ER), an intracellular site of Brucella replication. Possibly via binding of the ER chaperone BiP, VceC causes ER stress with concomitant expression of proinflammatory cytokines. Thus, induction of the unfolded protein response may represent a novel pathway by which host cells can detect pathogens deploying a T4SS.


Assuntos
Sistemas de Secreção Bacterianos , Brucella abortus/metabolismo , Brucella abortus/patogenicidade , Resposta a Proteínas não Dobradas , Fatores de Virulência/metabolismo , Animais , Citocinas/metabolismo , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/ultraestrutura , Chaperona BiP do Retículo Endoplasmático , Feminino , Células HeLa , Humanos , Mediadores da Inflamação/metabolismo , Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Virulência/toxicidade
10.
J Immunol ; 187(5): 2748-54, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21804020

RESUMO

Activation of caspase-1 leads to pyroptosis, a program of cell death characterized by cell lysis and inflammatory cytokine release. Caspase-1 activation triggered by multiple nucleotide-binding oligomerization domain-like receptors (NLRs; NLRC4, NLRP1b, or NLRP3) leads to loss of lysosomes via their fusion with the cell surface, or lysosome exocytosis. Active caspase-1 increased cellular membrane permeability and intracellular calcium levels, which facilitated lysosome exocytosis and release of host antimicrobial factors and microbial products. Lysosome exocytosis has been proposed to mediate secretion of IL-1ß and IL-18; however, blocking lysosome exocytosis did not alter cytokine processing or release. These studies indicate two conserved secretion pathways are initiated by caspase-1, lysosome exocytosis, and a parallel pathway resulting in cytokine release, and both enhance the antimicrobial nature of pyroptosis.


Assuntos
Apoptose/fisiologia , Caspase 1/metabolismo , Citocinas/metabolismo , Exocitose/fisiologia , Lisossomos/metabolismo , Macrófagos/metabolismo , Animais , Western Blotting , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
11.
Infect Immun ; 79(3): 1033-43, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21173315

RESUMO

As bacterial pathogens develop resistance against most currently used antibiotics, novel alternatives for treatment of microbial infectious diseases are urgently needed. Targeting bacterial virulence functions in order to disarm pathogens represents a promising alternative to classical antibiotic therapy. Type IV secretion systems, which are multiprotein complexes in the cell envelope that translocate effectors into host cells, are critical bacterial virulence factors in many pathogens and excellent targets for such "antivirulence" drugs. The VirB8 protein from the mammalian pathogen Brucella was chosen as a specific target, since it is an essential type IV secretion system component, it participates in multiple protein-protein interactions, and it is essential for the assembly of this translocation machinery. The bacterial two-hybrid system was adapted to assay VirB8 interactions, and a high-throughput screen identified specific small-molecule inhibitors. VirB8 interaction inhibitors also reduced the levels of VirB8 and of other VirB proteins, and many of them inhibited virB gene transcription in Brucella abortus 2308, suggesting that targeting of the secretion system has complex regulatory effects in vivo. One compound strongly inhibited the intracellular proliferation of B. abortus 2308 in a J774 macrophage infection model. The results presented here show that in vivo screens with the bacterial two-hybrid assay are suited to the identification of inhibitors of Brucella type IV secretion system function.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Sistemas de Secreção Bacterianos/efeitos dos fármacos , Brucella abortus/efeitos dos fármacos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Animais , Brucella abortus/crescimento & desenvolvimento , Brucella abortus/metabolismo , Linhagem Celular , Macrófagos/efeitos dos fármacos , Macrófagos/microbiologia , Camundongos , Viabilidade Microbiana , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Técnicas do Sistema de Duplo-Híbrido
12.
Mol Microbiol ; 70(6): 1378-96, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19019140

RESUMO

Survival and replication inside host cells by Brucella spp. requires a type IV secretion system (T4SS), encoded by the virB locus. However, the identity of the molecules secreted by the T4SS has remained elusive. We hypothesized that proteins translocated by the T4SS would be co-regulated with the virB operon. The LuxR family regulator VjbR, known to regulate virB, bound a fragment of the virB promoter containing an 18 bp palindromic motif (virB promoter box), showing that VjbR regulated the virB operon directly. To identify virB co-regulated genes, we searched the Brucella suis 1330 and B. abortus 2308 genomes for genes with an upstream virB promoter box. One hundred and forty-four promoters in the two genomes contained the virB promoter box, including those of fliC encoding flagellin and cgs encoding cyclic beta-glucan synthetase. Thirteen of these proteins were tested for VirB-dependent translocation into macrophages using a beta-lactamase reporter assay. This analysis resulted in the identification of the proteins encoded by BAB1_1652 (VceA) and BR1038/BAB1_1058 (VceC) as novel protein substrates of the Brucella T4SS. VceC could also be translocated by the Legionella pneumophila Dot/Icm T4SS into host cells. Our results suggest that VjbR co-ordinates expression of the T4SS and at least two of its secreted substrates.


Assuntos
Proteínas de Bactérias/metabolismo , Brucella abortus/metabolismo , Brucella suis/metabolismo , Macrófagos/metabolismo , Regulon , Via Secretória , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/genética , Sequência de Bases , Brucella abortus/genética , Brucella suis/genética , Linhagem Celular , Sequência Consenso , Ensaio de Desvio de Mobilidade Eletroforética , Escherichia coli/genética , Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Teste de Complementação Genética , Macrófagos/microbiologia , Camundongos , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Transporte Proteico , Via Secretória/genética
13.
J Bacteriol ; 190(13): 4427-36, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18469100

RESUMO

The Brucella abortus virB locus contains 12 open reading frames, termed virB1 through virB12, which encode a type IV secretion system. Polar mutations in the virB locus markedly reduce the ability of B. abortus to survive in cultured macrophages or to persist in organs of mice. While a nonpolar deletion of the virB2 gene reduces survival in cultured macrophages and in organs of mice, a nonpolar deletion of virB1 only reduces survival in macrophages, whereas virB12 is dispensable for either virulence trait. Here we investigated the role of the remaining genes in the virB locus during survival in macrophages and virulence in mice. Mutants carrying nonpolar deletions of the virB3, virB4, virB5, virB6, virB7, virB8, virB9, virB10, or virB11 gene were constructed and characterized. All mutations reduced the ability of B. abortus to survive in J774A.1 mouse macrophage-like cells to a degree similar to that caused by a deletion of the entire virB locus. Deletion of virB3, virB4, virB5, virB6, virB8, virB9, virB10, or virB11 markedly reduced the ability of B. abortus to persist in the spleens of mice at 8 weeks after infection. Interestingly, deletion of virB7 did not reduce the ability of B. abortus to persist in spleens of mice. We conclude that virB2, virB3, virB4, virB5, virB6, virB8, virB9, virB10, and virB11 are essential for virulence of B. abortus in mice, while functions encoded by the virB1, virB7, and virB12 genes are not required for persistence in organs with this animal model.


Assuntos
Proteínas de Bactérias/genética , Brucella/genética , Regulação Bacteriana da Expressão Gênica , Sistema Fagocitário Mononuclear/microbiologia , Animais , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/fisiologia , Western Blotting , Brucella/patogenicidade , Brucella abortus/genética , Brucella abortus/patogenicidade , Linhagem Celular , Feminino , Genes Bacterianos , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Óperon/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/microbiologia , Virulência/genética
14.
Infect Immun ; 73(9): 6048-54, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16113325

RESUMO

The Brucella abortus virB operon, consisting of 11 genes, virB1 to virB11, and two putative genes, orf12 (virB12) and orf13, encodes a type IV secretion system (T4SS) that is required for intracellular replication and persistent infection in the mouse model. This study was undertaken to determine whether orf12 (virB12) encodes an essential part of the T4SS apparatus. The virB12 gene was found to encode a 17-kDa protein, which was detected in vitro in B. abortus grown to stationary phase. Mice infected with B. abortus 2308 produced an antibody response to the protein encoded by virB12, showing that this gene is expressed during infection. Expression of virB12 was not required for survival in J774 macrophages. VirB12 was also dispensable for the persistence of B. abortus, B. melitensis, and B. suis in mice up to 4 weeks after infection, since deletion mutants lacking virB12 were recovered from splenic tissue at wild-type levels. These results show that VirB12 is not essential for the persistence of the human-pathogenic Brucella spp. in the mouse and macrophage models of infection.


Assuntos
Proteínas de Bactérias/biossíntese , Brucella abortus/genética , Brucelose/microbiologia , Regulação da Expressão Gênica/imunologia , Óperon , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , Brucella abortus/imunologia , Brucella abortus/metabolismo , Brucella melitensis/genética , Brucella suis/genética , Brucelose/metabolismo , Linhagem Celular , Feminino , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Deleção de Sequência , Baço/microbiologia
15.
Infect Immun ; 72(9): 5143-9, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15322008

RESUMO

The Brucella abortus virB operon, encoding a type IV secretion system (T4SS), is required for intracellular replication and persistent infection in the mouse model. The products of the first two genes of the virB operon, virB1 and virB2, are predicted to be localized at the bacterial surface, where they could potentially interact with host cells. Studies to date have focused on characterization of transposon mutations in these genes, which are expected to exert polar effects on downstream genes in the operon. In order to determine whether VirB1 and VirB2 are required for the function of the T4SS apparatus, we constructed and characterized nonpolar deletion mutations of virB1 and virB2. Both mutants were shown to be nonpolar, as demonstrated by their ability to express the downstream gene virB5 during stationary phase of growth in vitro. Both VirB1 and VirB2 were essential for intracellular replication in J774 macrophages. The nonpolar virB2 mutant was unable to cause persistent infection in the mouse model, demonstrating the essential role of VirB2 in the function of the T4SS apparatus during infection. In contrast, the nonpolar virB1 mutant persisted at wild-type levels, showing that the function of VirB1 is dispensable in the mouse model of persistent infection.


Assuntos
Proteínas de Bactérias/metabolismo , Brucella abortus/patogenicidade , Brucelose/microbiologia , Brucelose/fisiopatologia , Regulação Bacteriana da Expressão Gênica , Fatores de Virulência/metabolismo , Animais , Proteínas de Bactérias/genética , Brucella abortus/genética , Brucella abortus/crescimento & desenvolvimento , Brucella abortus/metabolismo , Linhagem Celular , Feminino , Deleção de Genes , Humanos , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Óperon , Baço/microbiologia , Virulência , Fatores de Virulência/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA