Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 16(10): e1009020, 2020 10.
Article in English | MEDLINE | ID: mdl-33108406

ABSTRACT

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.


Subject(s)
Brucella/metabolism , Brucellosis/metabolism , Membrane Proteins/biosynthesis , MicroRNAs/metabolism , Animals , Brucella/genetics , Brucellosis/genetics , Female , Host-Pathogen Interactions/immunology , Macrophages/immunology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , RNA, Messenger/genetics , Type IV Secretion Systems/genetics , Type IV Secretion Systems/metabolism
2.
Infect Immun ; 86(7)2018 07.
Article in English | MEDLINE | ID: mdl-29735518

ABSTRACT

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.


Subject(s)
Antigen Presentation , Brucella/immunology , Histocompatibility Antigens Class I/immunology , Lymphocyte Activation , Ovalbumin/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Animals , Antigens, Bacterial/immunology , Cross Reactions , Immune Evasion , Mice , Mice, Inbred C57BL , Peptide Fragments/immunology
3.
Infect Immun ; 84(12): 3458-3470, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27672085

ABSTRACT

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.


Subject(s)
Brucella/pathogenicity , Brucellosis/microbiology , Cyclic GMP/analogs & derivatives , Adaptation, Physiological , Animals , Biofilms , Brucella/metabolism , Brucella/ultrastructure , Brucellosis/pathology , Cells, Cultured , Cyclic GMP/genetics , Cyclic GMP/metabolism , Genetic Fitness , Macrophages/metabolism , Macrophages/microbiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mutation , Type IV Secretion Systems , Virulence
4.
PLoS Pathog ; 9(12): e1003785, 2013.
Article in English | MEDLINE | ID: mdl-24339776

ABSTRACT

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.


Subject(s)
Bacterial Proteins/physiology , Brucella melitensis/physiology , Macrophages/metabolism , Macrophages/microbiology , Unfolded Protein Response , Virulence Factors/physiology , Animals , Brucellosis/metabolism , Brucellosis/microbiology , Cells, Cultured , Dogs , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microbial Viability
5.
Biochem J ; 439(1): 79-83, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21692747

ABSTRACT

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.


Subject(s)
Bacterial Proteins/metabolism , Brucella melitensis/metabolism , Microtubules/metabolism , Receptors, Interleukin-1/metabolism , Bacterial Proteins/genetics , Brucella melitensis/genetics , Microtubules/drug effects , Nocodazole/pharmacology , Protein Structure, Tertiary , Receptors, Interleukin-1/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
6.
J Biol Chem ; 284(15): 9892-8, 2009 Apr 10.
Article in English | MEDLINE | ID: mdl-19196716

ABSTRACT

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.


Subject(s)
Bacterial Proteins/chemistry , Brucella/metabolism , Membrane Glycoproteins/chemistry , NF-kappa B/metabolism , Receptors, Interleukin-1/chemistry , Amino Acid Sequence , Animals , Bacterial Proteins/physiology , Cell Line , Cytoskeleton/metabolism , HeLa Cells , Humans , Membrane Glycoproteins/physiology , Mice , Molecular Sequence Data , Phosphatidylinositols/chemistry , Protein Structure, Tertiary , Receptors, Interleukin-1/physiology , Sequence Homology, Amino Acid
7.
J Immune Based Ther Vaccines ; 7: 1, 2009 Jan 06.
Article in English | MEDLINE | ID: mdl-19126207

ABSTRACT

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.

8.
Hum Gene Ther ; 16(1): 101-8, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15703493

ABSTRACT

Herpesvirus tegument protein VP22 has been shown to have biotherapeutic potential in tumor gene therapy. Some studies indicate that VP22 may enhance the transfer efficiency of therapeutic proteins by delivering them to more cells while trafficking. Our previous study showed that bovine herpesvirus VP22 (BVP22) enhanced equine herpesvirus thymidine kinase-ganciclovir (Etk-GCV) suicide gene therapy by an unknown intracellular effect. In this study, the interaction between BVP22 and host tumor cells was studied in neuroblastoma NXS2 cells. Cell cycle analysis was performed to determine whether BVP22 possesses biotherapeutic potential by altering the cell cycle, making cells more sensitive to therapeutic genes. As a result, the cell cycle was not affected by the transfection of BVP22 into NXS2 cells. However, cytotoxicity induced by BVP22 was observed in NXS2 cells on the second and third days after transient transfection. Further, analyses of caspase-3 activity and apoptosis suggested that BVP22 induces apoptosis in host tumor cells by upregulating the expression ratio of Bax to Bcl-2.


Subject(s)
Apoptosis , Cell Cycle , Neuroblastoma/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Viral Structural Proteins/pharmacology , Animals , Caspase 3 , Caspases/metabolism , Cattle , Humans , Mice , Neuroblastoma/pathology , Phosphoproteins/pharmacology , Sequence Deletion , Transfection , Tumor Cells, Cultured , Up-Regulation , Viral Structural Proteins/genetics , bcl-2-Associated X Protein
9.
Genet Vaccines Ther ; 2(1): 11, 2004 Aug 24.
Article in English | MEDLINE | ID: mdl-15327692

ABSTRACT

BACKGROUND: Viral promoters are used in mammalian expression vectors because they generally have strong activity in a wide variety of cells of differing tissues and species. METHODS: The utility of the BLV LTR/promoter (BLVp) for use in mammalian expression vectors was investigated through direct comparison to the CMV promoter (CMVp). Promoter activity was measured using luciferase assays of cell lines from different tissues and species stably transduced with BLVp or CMVp driven luciferase vectors including D17, FLK, BL3.1 and primary bovine B cells. Cells were also modified through the addition of BLV Tax expression vectors and/or BLV infection as well as treatment with trichostatin A (TSA). RESULTS: Results indicate the BLV promoter, while having low basal activity compared to the CMV promoter, can be induced to high-levels of activity similar to the CMV promoter in all cells tested. Tax or BLV infection specifically enhanced BLVp activity with no effect on CMVp activity. In contrast, the non-specific activator, TSA, enhanced both BLVp and CMVp activity. CONCLUSION: Based on these data, we conclude the BLV promoter could be very useful for transgene expression in mammalian expression vectors.

10.
J Virol ; 78(8): 4224-33, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15047837

ABSTRACT

Herpesvirus tegument protein VP22 can enhance the effect of therapeutic proteins in gene therapy, such as thymidine kinase (tk) and p53; however, the mechanism is unclear or controversial. In this study, mammalian expression vectors carrying bovine herpesvirus 1 (BHV-1) VP22 (BVP22) or herpes simplex virus type 1 (HSV-1) VP22 (HVP22) and equine herpesvirus type 4 (EHV-4) tk (Etk) were constructed in order to evaluate and compare the therapeutic potentials of BVP22 and HVP22 to enhance Etk/ganciclovir (Etk/GCV) suicide gene therapy for neuroblastomas by GCV cytotoxicity assays and noninvasive bioluminescent imaging in vitro and in vivo. BVP22 enhanced Etk/GCV cytotoxicity compared to that with HVP22 both in vitro and in vivo. However, assays utilizing a mixture of parental and stably transfected cells indicated that the enhancement was detected only in transfected cells. Thus, the therapeutic potential of BVP22 and HVP22 in Etk/GCV suicide gene therapy in this tumor system is not due to VP22 delivery of Etk into surrounding cells but rather is likely due to an enhanced intracellular effect.


Subject(s)
Genetic Therapy/methods , Neuroblastoma/therapy , Thymidine Kinase/genetics , Viral Structural Proteins/genetics , Animals , Artificial Gene Fusion , Cattle , Cell Line, Tumor , Ganciclovir/pharmacology , Genetic Vectors , Herpesvirus 4, Equid/enzymology , Herpesvirus 4, Equid/genetics , Humans , In Vitro Techniques , Mice , Transfection
11.
Curr Pharm Biotechnol ; 4(2): 99-107, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12678885

ABSTRACT

After 200 years of practice, vaccinology has proved to be very effective in preventing infectious diseases. However, several human and animal pathogens exist for which vaccines need to be improved or simply have not yet been discovered. The era of molecular genetic has given a new breath for vaccine development with the achievement of the "Third Generation of Vaccines": the DNA vaccine. In this article, we reviewed strategies that have been used to improve and modulate the immune response induced by DNA vaccines, using as a model the intracellular bacterial pathogen Brucella abortus. First, we described different approaches used to isolate and to identify genes that encode potential immunogens. Secondly, we reported the use of cytokine genes and genetic adjuvants that could improve the immunogenicity of target genes. And finally, we discussed the "Expression Library Immunization"-(ELI) strategy and the recent results obtained against Brucella abortus infection.


Subject(s)
Brucella abortus/drug effects , Intracellular Fluid/drug effects , Vaccines, DNA/administration & dosage , Animals , Brucella abortus/genetics , Humans , Intracellular Fluid/metabolism , Vaccines, DNA/genetics
12.
Viral Immunol ; 15(1): 155-63, 2002.
Article in English | MEDLINE | ID: mdl-11952137

ABSTRACT

Encephalomyocarditis virus (EMCV) and Mengo virus are highly virulent murine cardioviruses that are found in abundant quantities in the spleen and lymph nodes after infection. T lymphocytes are pivotal mediators of humoral and cellular immunity against cardioviral challenge, and are highly suspect candidates of EMCV and Mengo virus infection. We found T lymphocyte-like cell lines CTLL-2, EL-4, LY1+2/9, and LBRM33 were susceptible to productive viral infection and exhibited cytopathology after infection with virulent EMCV-R or attenuated Mengo virus strains vMC0 and vMC24. Flow cytometric analysis demonstrated progressive intracellular accumulation of viral proteins, such as the replication-dependent 3D viral polymerase, in EL-4 cells during infection. Conversely, freshly isolated and mitogen-stimulated CD4+ and CD8+ T cells were resistant to productive infection with these viruses, exhibiting no viral-induced cytopathic effects or intracellular presence of viral proteins. These data indicate that although T-lymphocyte-like tumor cell lines are highly susceptible to viral infection and cytopathic effects, primary/freshly isolated T cells are resistant to infection by EMCV-R or Mengo virus.


Subject(s)
Encephalomyocarditis virus/physiology , Mengovirus/physiology , T-Lymphocytes/virology , Animals , Cell Line , Cytopathogenic Effect, Viral , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Viral Proteins/analysis , Virus Replication , eIF-2 Kinase/physiology
13.
J Med Microbiol ; 51(1): 20-26, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11803949

ABSTRACT

The immunogenicity and protective efficacy of a DNA vaccine encoding the GroEL heat-shock gene from Brucella abortus was tested in BALB/c mice immunised by intramuscular (i.m.) needle injection or epidermally by gene gun. The Brucella GroEL gene was amplified by PCR and cloned into two different mammalian expression vectors pCMV-link and pCMV-tPA. The D17 cell line was transfected with both constructs and GroEL transcripts were detected by Northern blot. To determine the level of protein synthesised, transfected cell lysates were then submitted to Western blot. The non-secreted form of the recombinant GroEL produced by the pCMV-link construct was detected in much greater amount than the secreted form of the protein produced by the pCMV-tPA construct. After immunisation, a strong anti-GroEL IgG response was detected in mice vaccinated by i.m. injection or gene gun only when the pCMV-link/ GroEL plasmid was used. Regarding the pattern of immune response induced, i.m. needle injection raised a predominantly Th1 response with mostly IgG2a-specific anti-GroEL and high levels of IFN-gamma produced by splenic T cells. Gene gun immunisation induced a ThO type of immune response in mice characterised by a high IgG1/IgG2a ratio, and IL-4 and interferon (IFN)-gamma production. Even though a distinct pattern of immune response was generated depending upon the immunisation route used, neither method engendered a significant level of protection with the GroEL DNA vaccine.


Subject(s)
Brucella Vaccine/immunology , Brucella abortus/immunology , Chaperonin 60/immunology , Th1 Cells/immunology , Vaccines, DNA/immunology , Animals , Blotting, Northern , Blotting, Western , Brucella Vaccine/administration & dosage , Brucella Vaccine/genetics , Brucella abortus/genetics , Brucellosis/prevention & control , Cell Line , Chaperonin 60/genetics , DNA, Bacterial/immunology , Disease Models, Animal , Gene Amplification , Genetic Vectors , Heat-Shock Proteins , Immunoglobulin G/analysis , Immunoglobulin G/biosynthesis , Injections, Intramuscular , Interferon-gamma/biosynthesis , Interleukin-4/biosynthesis , Mice , Mice, Inbred BALB C , Polymerase Chain Reaction , Recombinant Proteins/immunology , Transfection , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...