Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 16(10): e1009020, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33108406

RESUMEN

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.


Asunto(s)
Brucella/metabolismo , Brucelosis/metabolismo , Proteínas de la Membrana/biosíntesis , MicroARNs/metabolismo , Animales , Brucella/genética , Brucelosis/genética , Femenino , Interacciones Huésped-Patógeno/inmunología , Macrófagos/inmunología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , ARN Mensajero/genética , Sistemas de Secreción Tipo IV/genética , Sistemas de Secreción Tipo IV/metabolismo
2.
Infect Immun ; 86(7)2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29735518

RESUMEN

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.


Asunto(s)
Presentación de Antígeno , Brucella/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Activación de Linfocitos , Ovalbúmina/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Animales , Antígenos Bacterianos/inmunología , Reacciones Cruzadas , Evasión Inmune , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/inmunología
3.
Infect Immun ; 84(12): 3458-3470, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27672085

RESUMEN

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.


Asunto(s)
Brucella/patogenicidad , Brucelosis/microbiología , GMP Cíclico/análogos & derivados , Adaptación Fisiológica , Animales , Biopelículas , Brucella/metabolismo , Brucella/ultraestructura , Brucelosis/patología , Células Cultivadas , GMP Cíclico/genética , GMP Cíclico/metabolismo , Aptitud Genética , Macrófagos/metabolismo , Macrófagos/microbiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Sistemas de Secreción Tipo IV , Virulencia
4.
Infect Immun ; 83(12): 4759-71, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26416901

RESUMEN

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.


Asunto(s)
Brucella melitensis/inmunología , Brucelosis/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Anergia Clonal , Memoria Inmunológica , Inmunidad Adaptativa , Traslado Adoptivo , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Brucella melitensis/patogenicidad , Brucelosis/genética , Brucelosis/microbiología , Brucelosis/patología , Linfocitos T CD4-Positivos/microbiología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD4-Positivos/trasplante , Linfocitos T CD8-positivos/microbiología , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/trasplante , Enfermedad Crónica , Femenino , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Interferón gamma/genética , Interferón gamma/inmunología , Recuento de Linfocitos , Ratones , Ratones Endogámicos BALB C , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Transducción de Señal , Proteína del Gen 3 de Activación de Linfocitos
5.
PLoS Pathog ; 9(12): e1003785, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24339776

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/fisiología , Brucella melitensis/fisiología , Macrófagos/metabolismo , Macrófagos/microbiología , Respuesta de Proteína Desplegada , Factores de Virulencia/fisiología , Animales , Brucelosis/metabolismo , Brucelosis/microbiología , Células Cultivadas , Perros , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Viabilidad Microbiana
6.
Biochem J ; 439(1): 79-83, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21692747

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Brucella melitensis/metabolismo , Microtúbulos/metabolismo , Receptores de Interleucina-1/metabolismo , Proteínas Bacterianas/genética , Brucella melitensis/genética , Microtúbulos/efectos de los fármacos , Nocodazol/farmacología , Estructura Terciaria de Proteína , Receptores de Interleucina-1/genética , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
7.
Infect Immun ; 78(1): 168-76, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19884330

RESUMEN

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.


Asunto(s)
Antígenos Bacterianos/metabolismo , Brucella melitensis/fisiología , Brucelosis/inmunología , Linfocitos T CD8-positivos/fisiología , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Afinidad de Anticuerpos , Epítopos , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/inmunología , Complejo Mayor de Histocompatibilidad , Ratones , Ratones Endogámicos BALB C , Unión Proteica
8.
Biochem Biophys Res Commun ; 397(1): 59-63, 2010 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-20471373

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Brucella melitensis/metabolismo , Factores de Virulencia/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Línea Celular , Clonación Molecular , Escherichia coli/genética , Proteínas de Unión a Maltosa , Ratones , FN-kappa B/antagonistas & inhibidores , Proteínas de Unión Periplasmáticas/genética , Proteínas de Unión Periplasmáticas/aislamiento & purificación , Proteínas de Unión Periplasmáticas/metabolismo , Fosfatidilinositoles/metabolismo , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Factores de Virulencia/genética , Factores de Virulencia/aislamiento & purificación
9.
Comp Immunol Microbiol Infect Dis ; 73: 101490, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33068875

RESUMEN

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.


Asunto(s)
Brucella melitensis , Brucelosis/inmunología , Hígado/inmunología , Bazo/inmunología , Enfermedad Aguda , Animales , Brucelosis/patología , Linfocitos T CD8-positivos/inmunología , Enfermedad Crónica , Modelos Animales de Enfermedad , Femenino , Hígado/patología , Ratones , Ratones Endogámicos BALB C , Microscopía/métodos , Bazo/patología
10.
J Immune Based Ther Vaccines ; 7: 1, 2009 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-19126207

RESUMEN

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.

11.
Virus Res ; 131(2): 152-9, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17949843

RESUMEN

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.


Asunto(s)
Begomovirus/fisiología , ADN Viral/metabolismo , Glycine max/virología , Proteínas de Movimiento Viral en Plantas/metabolismo , ADN de Cadena Simple/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Escherichia coli/genética , Expresión Génica , Unión Proteica , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo
12.
BMC Genomics ; 7: 91, 2006 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-16638145

RESUMEN

BACKGROUND: Comparative genomic hybridization can rapidly identify chromosomal regions that vary between organisms and tissues. This technique has been applied to detecting differences between normal and cancerous tissues in eukaryotes as well as genomic variability in microbial strains and species. The density of oligonucleotide probes available on current microarray platforms is particularly well-suited for comparisons of organisms with smaller genomes like bacteria and yeast where an entire genome can be assayed on a single microarray with high resolution. Available methods for analyzing these experiments typically confine analyses to data from pre-defined annotated genome features, such as entire genes. Many of these methods are ill suited for datasets with the number of measurements typical of high-density microarrays. RESULTS: We present an algorithm for analyzing microarray hybridization data to aid identification of regions that vary between an unsequenced genome and a sequenced reference genome. The program, CGHScan, uses an iterative random walk approach integrating multi-layered significance testing to detect these regions from comparative genomic hybridization data. The algorithm tolerates a high level of noise in measurements of individual probe intensities and is relatively insensitive to the choice of method for normalizing probe intensity values and identifying probes that differ between samples. When applied to comparative genomic hybridization data from a published experiment, CGHScan identified eight of nine known deletions in a Brucella ovis strain as compared to Brucella melitensis. The same result was obtained using two different normalization methods and two different scores to classify data for individual probes as representing conserved or variable genomic regions. The undetected region is a small (58 base pair) deletion that is below the resolution of CGHScan given the array design employed in the study. CONCLUSION: CGHScan is an effective tool for analyzing comparative genomic hybridization data from high-density microarrays. The algorithm is capable of accurately identifying known variable regions and is tolerant of high noise and varying methods of data preprocessing. Statistical analysis is used to define each variable region providing a robust and reliable method for rapid identification of genomic differences independent of annotated gene boundaries.


Asunto(s)
Algoritmos , Variación Genética , Genómica/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Brucella melitensis/genética , Brucella ovis/genética , Deleción Cromosómica , Genoma Bacteriano
13.
J Leukoc Biol ; 78(2): 401-11, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15857936

RESUMEN

Induction of immune responses against microbial antigens using DNA is an attractive strategy to mimic the immunity induced by live vaccines. Although DNA vaccines are efficacious in murine models, the requirement for multiple immunizations using high doses in outbred animals and humans has hindered deployment. This requirement is, in part, a result of poor vaccine spreading and suboptimal DC transfection efficiency. Incorporation of a signal that directs intercellular spreading of a DNA-encoded antigen is proposed to mimic live vaccine spreading and increase dendritic cell (DC) presentation. Bovine herpes virus 1 tegument protein, BVP22, is capable of trafficking to surrounding cells. To test the hypothesis that BVP22 enhances spreading and antigen presentation to CD4+ T cells, a DNA construct containing BVP22, fused in-frame to a sequence encoding a T cell epitope of Anaplasma marginale, was generated. A construct with reversed BVP22 sequence served as a negative control. Immunocytometric analysis of transfected primary keratinocytes, human embryonic kidney 293, COS-7, and Chinese hamster ovary cells showed that BVP22 enhanced intercellular spreading by > or = 150-fold. Flow cytometric analysis of antigen-presenting cells (APCs) positively selected from cocultures of transfected cells and APCs showed that 5% of test APCs were antigen-positive, compared with 0.6% of control APCs. Antigen-specific CD4+ T cell proliferation demonstrated that BVP22 enhanced DC antigen presentation by > or = 20-fold. This first report of the ability of BVP22 to increase DNA-encoded antigen acquisition by DCs and macrophages, with subsequent enhancement of major histocompatibility complex class II-restricted CD4+ T cell responses, supports incorporating a spreading motif in a DNA vaccine to target CD4+ T cell-dependent immunity in outbred animals.


Asunto(s)
Anaplasma marginale/inmunología , Vacunas Bacterianas/inmunología , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Epítopos de Linfocito T/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Vacunas de ADN/inmunología , Proteínas Estructurales Virales/inmunología , Animales , Presentación de Antígeno/inmunología , Vacunas Bacterianas/genética , Células CHO , Células COS , Bovinos , Chlorocebus aethiops , Técnicas de Cocultivo , Cricetinae , Cricetulus , Células Dendríticas/citología , Epítopos de Linfocito T/genética , Humanos , Queratinocitos/citología , Queratinocitos/inmunología , Macrófagos/citología , Macrófagos/inmunología , Ratones , Transporte de Proteínas/genética , Transporte de Proteínas/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Vacunas de ADN/genética , Proteínas Estructurales Virales/genética
14.
Adv Biochem Eng Biotechnol ; 154: 119-45, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-25395174

RESUMEN

: 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.


Asunto(s)
Bacterias/patogenicidad , Luminiscencia , Mediciones Luminiscentes , Animales , Luciferasas , Ratones , Enfermedades de las Plantas/microbiología , Plantas/microbiología
15.
Hum Gene Ther ; 16(1): 101-8, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15703493

RESUMEN

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.


Asunto(s)
Apoptosis , Ciclo Celular , Neuroblastoma/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Estructurales Virales/farmacología , Animales , Caspasa 3 , Caspasas/metabolismo , Bovinos , Humanos , Ratones , Neuroblastoma/patología , Fosfoproteínas/farmacología , Eliminación de Secuencia , Transfección , Células Tumorales Cultivadas , Regulación hacia Arriba , Proteínas Estructurales Virales/genética , Proteína X Asociada a bcl-2
16.
Viral Immunol ; 15(1): 155-63, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11952137

RESUMEN

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.


Asunto(s)
Virus de la Encefalomiocarditis/fisiología , Mengovirus/fisiología , Linfocitos T/virología , Animales , Línea Celular , Efecto Citopatogénico Viral , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Proteínas Virales/análisis , Replicación Viral , eIF-2 Quinasa/fisiología
17.
Genet Vaccines Ther ; 2(1): 11, 2004 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-15327692

RESUMEN

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.

18.
J Med Microbiol ; 51(8): 661-671, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12171297

RESUMEN

To identify antigens of Brucella spp. that are potentially involved in stimulating a protective T-cell-mediated immune response, previous studies identified 10 clones from a Brucella abortus 2308 genomic library with primed lymphocytes as probes. One selected positive clone (182) contained an insert of 1.2 kb which was identified, sequenced and characterised. The deduced amino acid sequence of the open reading frame (ORF) revealed 82% and 81% identity to the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) enzymes from Agrobacterium tumefaciens and Xanthobacter flavus, respectively. Southern blot analysis demonstrated that the gap gene is present in only one copy in the Brucella genome. B. abortus GAPDH was then expressed in Escherichia coli as a fusion protein with the maltose-binding protein (MBP). To demonstrate the functional activity of Brucella GAPDH, E. coli gap mutants were transformed with a Brucella pMAL-gap construct. Genetic complementation was achieved and as a result E. coli mutants were able to grow on glucose or other carbon source medium. The humoral and cellular immune responses to the recombinant (r) GAPDH were characterised. In Western blots, sera from naturally infected cattle and sheep showed antibody reactivity against rGAPDH. In response to in-vitro stimulation by rGAPDH, splenocytes from mice vaccinated with rGAPDH or B. abortus S19 were able to produce gamma-interferon and tumour necrosis factor-a but not interleukin (IL)-4. Furthermore, gap associated with murine IL-12 gene in a DNA vaccine formulation partially protected mice against experimental infection.


Asunto(s)
Vacuna contra la Brucelosis/inmunología , Brucella abortus/enzimología , Brucelosis Bovina/inmunología , Gliceraldehído-3-Fosfato Deshidrogenasas/inmunología , Interleucina-12/inmunología , Células TH1/inmunología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Brucella abortus/genética , Brucella abortus/inmunología , Brucelosis Bovina/microbiología , Brucelosis Bovina/prevención & control , Bovinos , ADN Bacteriano/química , ADN Bacteriano/genética , Prueba de Complementación Genética , Gliceraldehído-3-Fosfato Deshidrogenasas/genética , Interleucina-12/genética , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Datos de Secuencia Molecular , Plásmidos/genética , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Homología de Secuencia de Aminoácido , Vacunas de ADN/inmunología
19.
Genome Announc ; 2(3)2014 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-24874680

RESUMEN

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.

20.
Dis Model Mech ; 6(3): 811-8, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23519029

RESUMEN

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.


Asunto(s)
Huesos/microbiología , Huesos/patología , Brucelosis/microbiología , Brucelosis/patología , Articulaciones/microbiología , Articulaciones/patología , Animales , Artritis/microbiología , Artritis/patología , Médula Ósea/microbiología , Médula Ósea/patología , Brucella melitensis/fisiología , Femenino , Interacciones Huésped-Patógeno , Humanos , Hígado/microbiología , Hígado/patología , Ratones , Ratones Endogámicos BALB C , Bazo/microbiología , Bazo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA