Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Comp Immunol Microbiol Infect Dis ; 73: 101490, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33068875

RESUMO

Brucella melitensis is an intracellular bacteria causing disease in humans as an incidental host. The infection initiates as acute flu-like symptoms and may transform into a chronic cyclic infection. This cyclic infection may be partly due to the bacteria's ability to persist within antigen presenting cells and evade the CD8 + T cell response over long periods of time. This research aims to characterize the immune response of the acute and chronic forms of brucellosis in the murine liver and spleen. We also sought to determine if the exhaustion of the CD8 + T cells was a permanent or temporary change. This was accomplished by using adoptive transfer of acutely infected CD8 + T cells and chronically infected CD8 + T cells into a naïve host followed by re-infection. The histological examination presented supports the concept that exhausted T-cells can regain function through evidence of granulomatous inflammation after virulent challenge in a new host environment.


Assuntos
Brucella melitensis , Brucelose/imunologia , Fígado/imunologia , Baço/imunologia , Doença Aguda , Animais , Brucelose/patologia , Linfócitos T CD8-Positivos/imunologia , Doença Crônica , Modelos Animais de Doenças , Feminino , Fígado/patologia , Camundongos , Camundongos Endogâmicos BALB C , Microscopia/métodos , Baço/patologia
2.
PLoS Pathog ; 16(10): e1009020, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33108406

RESUMO

Brucellosis, caused by a number of Brucella species, remains the most prevalent zoonotic disease worldwide. Brucella establish chronic infections within host macrophages despite triggering cytosolic innate immune sensors, including Stimulator of Interferon Genes (STING), which potentially limit infection. In this study, STING was required for control of chronic Brucella infection in vivo. However, early during infection, Brucella down-regulated STING mRNA and protein. Down-regulation occurred post-transcriptionally, required live bacteria, the Brucella type IV secretion system, and was independent of host IRE1-RNase activity. STING suppression occurred in MyD88-/- macrophages and was not induced by Toll-like receptor agonists or purified Brucella lipopolysaccharide (LPS). Rather, Brucella induced a STING-targeting microRNA, miR-24-2, in a type IV secretion system-dependent manner. Furthermore, STING downregulation was inhibited by miR-24 anti-miRs and in Mirn23a locus-deficient macrophages. Failure to suppress STING expression in Mirn23a-/- macrophages correlated with diminished Brucella replication, and was rescued by exogenous miR-24. Mirn23a-/- mice were also more resistant to splenic colonization one week post infection. Anti-miR-24 potently suppressed replication in wild type, but much less in STING-/- macrophages, suggesting most of the impact of miR-24 induction on replication occurred via STING suppression. In summary, Brucella sabotages cytosolic surveillance by miR-24-dependent suppression of STING expression; post-STING activation "damage control" via targeted STING destruction may enable establishment of chronic infection.


Assuntos
Brucella/metabolismo , Brucelose/metabolismo , Proteínas de Membrana/biossíntese , MicroRNAs/metabolismo , Animais , Brucella/genética , Brucelose/genética , Feminino , Interações Hospedeiro-Patógeno/imunologia , Macrófagos/imunologia , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , RNA Mensageiro/genética , Sistemas de Secreção Tipo IV/genética , Sistemas de Secreção Tipo IV/metabolismo
3.
Infect Immun ; 86(7)2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29735518

RESUMO

Brucella spp. are intracellular pathogenic bacteria remarkable in their ability to escape immune surveillance and therefore inflict a state of chronic disease within the host. To enable further immune response studies, Brucella was engineered to express the well-characterized chicken ovalbumin (OVA). Surprisingly, we found that CD8 T cells bearing T cell receptors (TCR) nominally specific for the OVA peptide SIINFEKL (OT-1) reacted to parental Brucella-infected targets as well as OVA-expressing Brucella variants in cytotoxicity assays. Furthermore, splenocytes from Brucella-immunized mice produced gamma interferon (IFN-γ) and exhibited cytotoxicity in response to SIINFEKL-pulsed target cells.To determine if the SIINFEKL-reactive OT-1 TCR could be cross-reacting to Brucella peptides, we searched the Brucella proteome using an algorithm to generate a list of near-neighbor nonamer peptides that would bind to H2Kb Selecting five Brucella peptide candidates, along with controls, we verified that several of these peptides mimicked SIINFEKL, resulting in T cell activation through the "SIINFEKL-specific" TCR. Activation was dependent on peptide concentration as well as sequence. Our results underscore the complexity and ubiquity of cross-reactivity in T cell recognition. This cross-reactivity may enable microbes such as Brucella to escape immune surveillance by presenting peptides similar to those of the host and may also lead to the activation of autoreactive T cells.


Assuntos
Apresentação de Antígeno , Brucella/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Ativação Linfocitária , Ovalbumina/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Animais , Antígenos de Bactérias/imunologia , Reações Cruzadas , Evasão da Resposta Imune , Camundongos , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos/imunologia
4.
Infect Immun ; 84(12): 3458-3470, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27672085

RESUMO

Brucella species are facultative intracellular bacteria that cause brucellosis, a chronic debilitating disease significantly impacting global health and prosperity. Much remains to be learned about how Brucella spp. succeed in sabotaging immune host cells and how Brucella spp. respond to environmental challenges. Multiple types of bacteria employ the prokaryotic second messenger cyclic di-GMP (c-di-GMP) to coordinate responses to shifting environments. To determine the role of c-di-GMP in Brucella physiology and in shaping host-Brucella interactions, we utilized c-di-GMP regulatory enzyme deletion mutants. Our results show that a ΔbpdA phosphodiesterase mutant producing excess c-di-GMP displays marked attenuation in vitro and in vivo during later infections. Although c-di-GMP is known to stimulate the innate sensor STING, surprisingly, the ΔbpdA mutant induced a weaker host immune response than did wild-type Brucella or the low-c-di-GMP guanylate cyclase ΔcgsB mutant. Proteomics analysis revealed that c-di-GMP regulates several processes critical for virulence, including cell wall and biofilm formation, nutrient acquisition, and the type IV secretion system. Finally, ΔbpdA mutants exhibited altered morphology and were hypersensitive to nutrient-limiting conditions. In summary, our results indicate a vital role for c-di-GMP in allowing Brucella to successfully navigate stressful and shifting environments to establish intracellular infection.


Assuntos
Brucella/patogenicidade , Brucelose/microbiologia , GMP Cíclico/análogos & derivados , Adaptação Fisiológica , Animais , Biofilmes , Brucella/metabolismo , Brucella/ultraestrutura , Brucelose/patologia , Células Cultivadas , GMP Cíclico/genética , GMP Cíclico/metabolismo , Aptidão Genética , Macrófagos/metabolismo , Macrófagos/microbiologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Sistemas de Secreção Tipo IV , Virulência
5.
Adv Biochem Eng Biotechnol ; 154: 119-45, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-25395174

RESUMO

: Bioluminescence imaging (BLI) of bacteria was primarily designed to permit real-time, sensitive, and noninvasive monitoring of the progression of infection in live animals. Generally, BLI relies on the construction of bacterial strains that possess the lux operon. The lux operon is composed of a set of genes that encode the luciferase enzyme and its cognate substrate, which interact to produce light-a phenomenon that is referred to as bioluminescence. Bioluminescence emitted by the bacteria can then be detected and imaged within a living host using sensitive charge-coupled device (CCD) cameras. In comparison to traditional host-pathogen studies, BLI offers the opportunity for extended monitoring of infected animals without resorting to euthanasia and extensive tissue processing at each time point. Therefore, BLI can reduce the number of animals required to generate meaningful data, while significantly contributing to the understanding of pathogenesis in the host and, subsequently, the development and evaluation of adequate vaccines and therapeutics. BLI is also useful in characterizing the interactions of pathogens with plants and the para-host environment. In this chapter, we demonstrate the broad application of BLI for studying bacterial pathogens in different niches. Furthermore, we will specifically focus on the use of BLI to characterize the following: (1) the pathogenesis of Brucella melitensis in mice (animal host), and (2) the progression of infection of Clavibacter michiganensis subsp. michiganensis in tomatoes (plant host). These studies will provide an overview of the wide potential of BLI and its role in enhancing the study of unique-and sometimes difficult-to-characterize-bacterial pathogens.


Assuntos
Bactérias/patogenicidade , Luminescência , Medições Luminescentes , Animais , Luciferases , Camundongos , Doenças das Plantas/microbiologia , Plantas/microbiologia
6.
Infect Immun ; 83(12): 4759-71, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26416901

RESUMO

Brucella melitensis is a well-adapted zoonotic pathogen considered a scourge of mankind since recorded history. In some cases, initial infection leads to chronic and reactivating brucellosis, incurring significant morbidity and economic loss. The mechanism by which B. melitensis subverts adaptive immunological memory is poorly understood. Previous work has shown that Brucella-specific CD8(+) T cells express gamma interferon (IFN-γ) and can transition to long-lived memory cells but are not polyfunctional. In this study, chronic infection of mice with B. melitensis led to CD8(+) T cell exhaustion, manifested by programmed cell death 1 (PD-1) and lymphocyte activation gene 3 (LAG-3) expression and a lack of IFN-γ production. The B. melitensis-specific CD8(+) T cells that produced IFN-γ expressed less IFN-γ per cell than did CD8(+) cells from uninfected mice. Both memory precursor (CD8(+) LFA1(HI) CD127(HI) KLRG1(LO)) and long-lived memory (CD8(+) CD27(HI) CD127(HI) KLRG1(LO)) cells were identified during chronic infection. Interestingly, after adoptive transfer, mice receiving cells from chronically infected animals were able to contain infection more rapidly than recipients of cells from acutely infected or uninfected donors, although the proportions of exhausted CD8(+) T cells increased after adoptive transfer in both challenged and unchallenged recipients. CD8(+) T cells of challenged recipients initially retained the stunted IFN-γ production found prior to transfer, and cells from acutely infected mice were never seen to transition to either memory subset at all time points tested, up to 30 days post-primary infection, suggesting a delay in the generation of memory. Here we have identified defects in Brucella-responsive CD8(+) T cells that allow chronic persistence of infection.


Assuntos
Brucella melitensis/imunologia , Brucelose/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Anergia Clonal , Memória Imunológica , Imunidade Adaptativa , Transferência Adotiva , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Brucella melitensis/patogenicidade , Brucelose/genética , Brucelose/microbiologia , Brucelose/patologia , Linfócitos T CD4-Positivos/microbiologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD4-Positivos/transplante , Linfócitos T CD8-Positivos/microbiologia , Linfócitos T CD8-Positivos/patologia , Linfócitos T CD8-Positivos/transplante , Doença Crônica , Feminino , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Interferon gama/genética , Interferon gama/imunologia , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/imunologia , Transdução de Sinais , Proteína do Gene 3 de Ativação de Linfócitos
7.
Genome Announc ; 2(3)2014 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-24874680

RESUMO

Brucella spp. are facultative intracellular bacterial pathogens causing the zoonotic disease brucellosis. Here, we report the draft genome sequence of the Brucella melitensis strain from India designated Bm IND1, isolated from stomach contents of an aborted goat fetus.

8.
PLoS Pathog ; 9(12): e1003785, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24339776

RESUMO

Brucella melitensis is a facultative intracellular bacterium that causes brucellosis, the most prevalent zoonosis worldwide. The Brucella intracellular replicative niche in macrophages and dendritic cells thwarts immune surveillance and complicates both therapy and vaccine development. Currently, host-pathogen interactions supporting Brucella replication are poorly understood. Brucella fuses with the endoplasmic reticulum (ER) to replicate, resulting in dramatic restructuring of the ER. This ER disruption raises the possibility that Brucella provokes an ER stress response called the Unfolded Protein Response (UPR). In this study, B. melitensis infection up regulated expression of the UPR target genes BiP, CHOP, and ERdj4, and induced XBP1 mRNA splicing in murine macrophages. These data implicate activation of all 3 major signaling pathways of the UPR. Consistent with previous reports, XBP1 mRNA splicing was largely MyD88-dependent. However, up regulation of CHOP, and ERdj4 was completely MyD88 independent. Heat killed Brucella stimulated significantly less BiP, CHOP, and ERdj4 expression, but induced XBP1 splicing. Although a Brucella VirB mutant showed relatively intact UPR induction, a TcpB mutant had significantly compromised BiP, CHOP and ERdj4 expression. Purified TcpB, a protein recently identified to modulate microtubules in a manner similar to paclitaxel, also induced UPR target gene expression and resulted in dramatic restructuring of the ER. In contrast, infection with the TcpB mutant resulted in much less ER structural disruption. Finally, tauroursodeoxycholic acid, a pharmacologic chaperone that ameliorates the UPR, significantly impaired Brucella replication in macrophages. Together, these results suggest Brucella induces a UPR, via TcpB and potentially other factors, that enables its intracellular replication. Thus, the UPR may provide a novel therapeutic target for the treatment of brucellosis. These results also have implications for other intracellular bacteria that rely on host physiologic stress responses for replication.


Assuntos
Proteínas de Bactérias/fisiologia , Brucella melitensis/fisiologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Resposta a Proteínas não Dobradas , Fatores de Virulência/fisiologia , Animais , Brucelose/metabolismo , Brucelose/microbiologia , Células Cultivadas , Cães , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Viabilidade Microbiana
9.
Dis Model Mech ; 6(3): 811-8, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23519029

RESUMO

Brucellosis, a frequent bacterial zoonosis, can produce debilitating chronic disease with involvement of multiple organs in human patients. Whereas acute brucellosis is well studied using the murine animal model, long-term complications of host-pathogen interaction remain largely elusive. Human brucellosis frequently results in persistent, chronic osteoarticular system involvement, with complications such as arthritis, spondylitis and sacroiliitis. Here, we focused on identifying infectious sites in the mouse that parallel Brucella melitensis foci observed in patients. In vivo imaging showed rapid bacterial dispersal to multiple sites of the murine axial skeleton. In agreement with these findings, immunohistochemistry revealed the presence of bacteria in bones and limbs, and in the lower spine vertebrae of the axial skeleton where they were preferentially located in the bone marrow. Surprisingly, some animals developed arthritis in paws and spine after infection, but without obvious bacteria in these sites. The identification of Brucella in the bones of mice corroborates the findings in humans that these osteoarticular sites are important niches for the persistence of Brucella in the host, but the mechanisms that mediate pathological manifestations in these sites remain unclear. Future studies addressing the immune responses within osteoarticular tissue foci could elucidate important tissue injury mediators and Brucella survival strategies.


Assuntos
Osso e Ossos/microbiologia , Osso e Ossos/patologia , Brucelose/microbiologia , Brucelose/patologia , Articulações/microbiologia , Articulações/patologia , Animais , Artrite/microbiologia , Artrite/patologia , Medula Óssea/microbiologia , Medula Óssea/patologia , Brucella melitensis/fisiologia , Feminino , Interações Hospedeiro-Patógeno , Humanos , Fígado/microbiologia , Fígado/patologia , Camundongos , Camundongos Endogâmicos BALB C , Baço/microbiologia , Baço/patologia
10.
PLoS One ; 7(4): e34925, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22558103

RESUMO

Brucellosis is a common zoonotic disease that remains endemic in many parts of the world. Dissecting the host immune response during this disease provides insight as to why brucellosis is often difficult to resolve. We used a Brucella epitope specific in vivo killing assay to investigate the ability of CD8+ T cells to kill targets treated with purified pathogenic protein. Importantly, we found the pathogenic protein TcpB to be a novel effector of adaptive immune evasion by inhibiting CD8+ T cell killing of Brucella epitope specific target cells in mice. Further, BALB/c mice show active Brucella melitensis infection beyond one year, many with previously unreported focal infection of the urogenital area. A fraction of CD8+ T cells show a CD8+ Tmem phenotype of LFA-1hi, CD127hi, KLRG-1lo during the course of chronic brucellosis, while the CD8+ T cell pool as a whole had a very weak polyfunctional cytokine response with diminished co-expression of IFN-γ with TNFα and/or IL-2, a hallmark of exhaustion. When investigating the expression of these 3 cytokines individually, we observed significant IFN-γ expression at 90 and 180 days post-infection. TNFα expression did not significantly exceed or fall below background levels at any time. IL-2 expression did not significantly exceeded background, but, interestingly, did fall significantly below that of uninfected mice at 180 days post-infection. Brucella melitensis evades and blunts adaptive immunity during acute infection and our findings provide potential mechanisms for the deficit observed in responding CD8+ T cells during chronic brucellosis.


Assuntos
Imunidade Adaptativa/imunologia , Brucelose/imunologia , Brucelose/fisiopatologia , Proteínas de Fímbrias/imunologia , Evasão da Resposta Imune/imunologia , Linfócitos T Citotóxicos/imunologia , Análise de Variância , Animais , Citocinas/metabolismo , Feminino , Citometria de Fluxo , Imunofenotipagem , Interferon gama/metabolismo , Interleucina-2/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Linfócitos T Citotóxicos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
11.
Biomol Concepts ; 3(6): 571-580, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23585820

RESUMO

The eukaryotic cytoskeleton is a vulnerable target of many microbial pathogens during the course of infection. Rearrangements of host cytoskeleton benefit microbes in various stages of their infection cycle such as invasion, motility, and persistence. Bacterial pathogens deliver a number of effector proteins into host cells for modulating the dynamics of actin and microtubule cytoskeleton. Alteration of the actin cytoskeleton is generally achieved by bacterial effectors that target the small GTPases of the host. Modulation of microtubule dynamics involves direct interaction of effector proteins with the subunits of microtubules or recruiting cellular proteins that affect microtubule dynamics. This review will discuss effector proteins from animal and human bacterial pathogens that either destabilize or stabilize host micro-tubules to advance the infectious process. A compilation of these research findings will provide an overview of known and unknown strategies used by various bacterial effectors to modulate the host microtubule dynamics. The present review will undoubtedly help direct future research to determine the mechanisms of action of many bacterial effector proteins and contribute to understanding the survival strategies of diverse adherent and invasive bacterial pathogens.

12.
Biochem J ; 439(1): 79-83, 2011 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-21692747

RESUMO

TIR (Toll/interleukin-1 receptor) domain-containing proteins play a crucial role in innate immunity in eukaryotes. Brucella is a highly infectious intracellular bacterium that encodes a TIR domain protein (TcpB) to subvert host innate immune responses to establish a beneficial niche for pathogenesis. TcpB inhibits NF-κB (nuclear factor κB) activation and pro-inflammatory cytokine secretions mediated by TLR (Toll-like receptor) 2 and TLR4. In the present study, we have demonstrated that TcpB modulates microtubule dynamics by acting as a stabilization factor. TcpB increased the rate of nucleation as well as the polymerization phases of microtubule formation in a similar manner to paclitaxel. TcpB could efficiently inhibit nocodazole- or cold-induced microtubule disassembly. Microtubule stabilization by TcpB is attributed to the BB-loop region of the TIR domain, and a point mutation affected the microtubule stabilization as well as the TLR-suppression properties of TcpB.


Assuntos
Proteínas de Bactérias/metabolismo , Brucella melitensis/metabolismo , Microtúbulos/metabolismo , Receptores de Interleucina-1/metabolismo , Proteínas de Bactérias/genética , Brucella melitensis/genética , Microtúbulos/efeitos dos fármacos , Nocodazol/farmacologia , Estrutura Terciária de Proteína , Receptores de Interleucina-1/genética , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
13.
Biochem Biophys Res Commun ; 397(1): 59-63, 2010 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-20471373

RESUMO

Toll/interleukin-1 like receptors are evolutionarily conserved proteins in eukaryotes that play crucial role in pathogen recognition and innate immune responses. Brucella are facultative intracellular bacterial pathogens causing brucellosis in animal and human hosts. Brucella behave as a stealthy pathogen by evading the immune recognition or suppressing the TLR signaling cascades. Brucella encode a TIR domain containing protein, TcpB, which suppresses NF-kappaB activation as well as pro-inflammatory cytokine secretion mediated by TLR2 and TLR4 receptors. TcpB targets the TIRAP mediated pathway to suppress TLR signaling. With the objective of detailed characterization, we have over expressed and purified TcpB from Brucella melitensis in native condition. The purified protein exhibited lipid-binding properties and cell permeability. NF-kappaB inhibition property of endogenous TcpB has also been demonstrated. The data provide insight into the mechanism of action of TcpB in the intracellular niche of Brucella.


Assuntos
Proteínas de Bactérias/metabolismo , Brucella melitensis/metabolismo , Fatores de Virulência/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/isolamento & purificação , Linhagem Celular , Clonagem Molecular , Escherichia coli/genética , Proteínas Ligantes de Maltose , Camundongos , NF-kappa B/antagonistas & inibidores , Proteínas Periplásmicas de Ligação/genética , Proteínas Periplásmicas de Ligação/isolamento & purificação , Proteínas Periplásmicas de Ligação/metabolismo , Fosfatidilinositóis/metabolismo , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/isolamento & purificação
14.
Infect Immun ; 78(1): 168-76, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19884330

RESUMO

Brucella spp. are intracellular bacteria that cause the most frequent zoonosis in the world. Although recent work has advanced the field of Brucella vaccine development, there remains no safe human vaccine. In order to produce a safe and effective human vaccine, the immune response to Brucella spp. requires greater understanding. Induction of Brucella-specific CD8+ T cells is considered an important aspect of the host response; however, the CD8+ T-cell response is not clearly defined. Discovering the epitope containing antigens recognized by Brucella-specific CD8+ T cells and correlating them with microarray data will aid in determining proteins critical for vaccine development that cover a kinetic continuum during infection. Developing tools to take advantage of the BALB/c mouse model of Brucella melitensis infection will help to clarify the correlates of immunity and improve the efficacy of this model. Two H-2(d) CD8+ T-cell epitopes have been characterized, and a group of immunogenic proteins have provoked gamma interferon production by CD8+ T cells. RYCINSASL and NGSSSMATV induced cognate CD8+ T cells after peptide immunization that showed specific killing in vivo. Importantly, we found by microarray analysis that the genes encoding these epitopes are differentially expressed following macrophage infection, further emphasizing that these discordant genes may play an important role in the pathogenesis of B. melitensis infection.


Assuntos
Antígenos de Bactérias/metabolismo , Brucella melitensis/fisiologia , Brucelose/imunologia , Linfócitos T CD8-Positivos/fisiologia , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/imunologia , Afinidade de Anticorpos , Epitopos , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/imunologia , Complexo Principal de Histocompatibilidade , Camundongos , Camundongos Endogâmicos BALB C , Ligação Proteica
15.
Trends Microbiol ; 17(8): 371-7, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19660951

RESUMO

Brucella is an important zoonotic pathogen for which no human vaccine exists. In an infected host, Brucella resides in macrophages but must coordinate expression of multiple virulence factors for successful cell entry and trafficking to acquire this replicative niche. Brucella responds to environmental signals to regulate virulence strategies that circumvent or blunt the host immune response. The Brucella quorum sensing system is a nexus of control for several Brucella virulence factors including flagellar genes and the type IV secretion system. Other sensory transduction systems, such as BvrRS and the newly described LOV-HK, sense environmental factors to control virulence. Here, we examine the contributions of various regulatory systems to Brucella virulence.


Assuntos
Brucella/fisiologia , Regulação Bacteriana da Expressão Gênica , Transdução de Sinais , Fatores de Virulência/biossíntese , Proteínas de Bactérias/biossíntese , Humanos , Percepção de Quorum , Virulência
16.
J Biol Chem ; 284(15): 9892-8, 2009 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-19196716

RESUMO

Toll-like receptors (TLRs) play essential roles in the activation of innate immune responses against microbial infections. TLRs and downstream adaptor molecules contain a conserved cytoplasmic TIR domain. TIRAP is a TIR domain-containing adaptor protein that recruits the signaling adaptor MyD88 to a subset of TLRs. Many pathogenic microorganisms subvert TLR signaling pathways to suppress host immune responses to benefit their survival and persistence. Brucella encodes a TIR domain-containing protein (TcpB) that inhibits TLR2- and TLR4-mediated NF-kappaB activation. Sequence analysis indicated a moderate level of similarity between TcpB and the TLR adaptor molecule TIRAP. We found that TcpB could efficiently block TIRAP-induced NF-kappaB activation. Subsequent studies revealed that by analogy to TIRAP, TcpB interacts with phosphoinositides through its N-terminal domain and colocalizes with the plasma membrane and components of the cytoskeleton. Our findings suggest that TcpB targets the TIRAP-mediated pathway to subvert TLR signaling. In vivo mouse studies indicated that TcpB-deficient Brucella is defective in systemic spread at the early stages of infection.


Assuntos
Proteínas de Bactérias/química , Brucella/metabolismo , Glicoproteínas de Membrana/química , NF-kappa B/metabolismo , Receptores de Interleucina-1/química , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/fisiologia , Linhagem Celular , Citoesqueleto/metabolismo , Células HeLa , Humanos , Glicoproteínas de Membrana/fisiologia , Camundongos , Dados de Sequência Molecular , Fosfatidilinositóis/química , Estrutura Terciária de Proteína , Receptores de Interleucina-1/fisiologia , Homologia de Sequência de Aminoácidos
17.
J Immune Based Ther Vaccines ; 7: 1, 2009 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-19126207

RESUMO

BACKGROUND: There is no safe, effective human vaccine against brucellosis. Live attenuated Brucella strains are widely used to vaccinate animals. However these live Brucella vaccines can cause disease and are unsafe for humans. Killed Brucella or subunit vaccines are not effective in eliciting long term protection. In this study, we evaluate an approach using a live, non-pathogenic bacteria (E. coli) genetically engineered to mimic the brucellae pathway of infection and present antigens for an appropriate cytolitic T cell response. METHODS: E. coli was modified to express invasin of Yersinia and listerialysin O (LLO) of Listeria to impart the necessary infectivity and antigen releasing traits of the intracellular pathogen, Brucella. This modified E. coli was considered our vaccine delivery system and was engineered to express Green Fluorescent Protein (GFP) or Brucella antigens for in vitro and in vivo immunological studies including cytokine profiling and cytotoxicity assays. RESULTS: The E. coli vaccine vector was able to infect all cells tested and efficiently deliver therapeutics to the host cell. Using GFP as antigen, we demonstrate that the E. coli vaccine vector elicits a Th1 cytokine profile in both primary and secondary immune responses. Additionally, using this vector to deliver a Brucella antigen, we demonstrate the ability of the E. coli vaccine vector to induce specific Cytotoxic T Lymphocytes (CTLs). CONCLUSION: Protection against most intracellular bacterial pathogens can be obtained mostly through cell mediated immunity. Data presented here suggest modified E. coli can be used as a vaccine vector for delivery of antigens and therapeutics mimicking the infection of the pathogen and inducing cell mediated immunity to that pathogen.

18.
Virus Res ; 131(2): 152-9, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17949843

RESUMO

Bipartite geminiviruses possess two movement proteins (NSP and MP), which mediate the intra- and intercellular movement. In order to accomplish the transport process the movement proteins interact with viral nucleic acids in a sequence non-specific manner. To investigate the nucleic acid recognition properties of MP of MYMIV-Sb, the protein was expressed in Escherichia coli as a fusion protein with maltose-binding protein (MBP) and purified in native condition. Gel mobility shift assay was employed for analyzing the DNA recognition properties of purified MBP-MP fusion protein. The analyses demonstrated the sequence non-specific binding of MYMIV-Sb MP to both ds and ssDNA and its high affinity for ssDNA. MP of MYMIV-Sb did not show any specificity towards various forms of plasmid DNA but displayed size selection towards linear dsDNA.


Assuntos
Begomovirus/fisiologia , DNA Viral/metabolismo , Glycine max/virologia , Proteínas do Movimento Viral em Plantas/metabolismo , DNA de Cadeia Simples/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Escherichia coli/genética , Expressão Gênica , Ligação Proteica , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo
19.
Science ; 317(5841): 1090-3, 2007 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-17717187

RESUMO

Histidine kinases, used for environmental sensing by bacterial two-component systems, are involved in regulation of bacterial gene expression, chemotaxis, phototaxis, and virulence. Flavin-containing domains function as light-sensory modules in plant and algal phototropins and in fungal blue-light receptors. We have discovered that the prokaryotes Brucella melitensis, Brucella abortus, Erythrobacter litoralis, and Pseudomonas syringae contain light-activated histidine kinases that bind a flavin chromophore and undergo photochemistry indicative of cysteinyl-flavin adduct formation. Infection of macrophages by B. abortus was stimulated by light in the wild type but was limited in photochemically inactive and null mutants, indicating that the flavin-containing histidine kinase functions as a photoreceptor regulating B. abortus virulence.


Assuntos
Brucella abortus/enzimologia , Brucella melitensis/enzimologia , Luz , Macrófagos/microbiologia , Proteínas Quinases/metabolismo , Pseudomonas syringae/enzimologia , Sphingomonadaceae/enzimologia , Trifosfato de Adenosina/metabolismo , Animais , Brucella abortus/crescimento & desenvolvimento , Brucella abortus/patogenicidade , Linhagem Celular , Clonagem Molecular , Ativação Enzimática , Mononucleotídeo de Flavina/metabolismo , Histidina Quinase , Camundongos , Dados de Sequência Molecular , Mutação , Fosforilação , Fotoquímica , Proteínas Quinases/química , Proteínas Quinases/genética , Estrutura Terciária de Proteína , Transdução de Sinais , Virulência
20.
Clin Vaccine Immunol ; 14(3): 304-11, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17215335

RESUMO

We developed a vaccine construct in which a BVP22 domain and an invariant-chain major histocompatibility complex class II-targeting motif capable of enhancing dendritic cell antigen uptake and presentation were fused to a sequence encoding a B- and T-cell antigen from the Anaplasma marginale major surface protein 1a and tested whether this construct would prime and expand immune responses in outbred calves. A single inoculation with this construct effectively primed the immune responses, as demonstrated by a significant enhancement of CD4(+) T-cell proliferation compared to that in calves identically inoculated but inoculated with a DNA construct lacking the targeting domains and compared to that in calves inoculated with an empty vector. These proliferative responses were mirrored by priming and expansion of gamma interferon-positive CD4(+) T cells and immunoglobulin G responses against the linked B-cell epitope. Priming by the single immunization induced memory that underwent rapid recall following reexposure to the antigen. These results demonstrate that DNA vaccines targeting key intercellular and intracellular events significantly enhance priming and expansion and support the feasibility of single-dose DNA immunization in outbred populations.


Assuntos
Antígenos de Diferenciação de Linfócitos B/imunologia , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Memória Imunológica , Vacinas de DNA/imunologia , Animais , Apresentação de Antígeno , Bovinos , Células Dendríticas/imunologia , Imunização , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA