Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Antioxidants (Basel) ; 12(10)2023 Oct 15.
Article in English | MEDLINE | ID: mdl-37891942

ABSTRACT

Enhanced solvent extraction (ESE) and pressurized liquid extraction (PLE) have been used for the first time to obtain antioxidant compounds from Prestonia mollis leaves. The effects of pressure (100-250 bar), temperature (55-75 °C) and the composition of the extraction solvent (ethanol, water and hydroalcoholic mixtures) were evaluated according to multilevel factorial designs. PLE provided the largest extraction yields compared to ESE, as well as a greater impact of the operating conditions studied. The highest total phenolic content was obtained when using a hydroalcoholic mixture (CO2/ethanol/water 50/25/25) through ESE at 100 bar and 75 °C. The antioxidant capacity of this extract is related to higher concentration levels of the identified flavonoids: Quercetin 3-O-xylosyl-rutinoside, Kaempferol 3-(2G-apiosylrobinobioside) and Kaempferol 4'-glucoside 7-rhamnoside. This extract was tested for the supercritical impregnation of polylactic acid (PLA), which is a polymer widely used in the biomedical industry. The influence of pressure (100-400 bar), temperature (35-55 °C), amount of extract (3-6 mL) and impregnation time (1-2 h) have been evaluated. The best results were obtained by impregnating 3 mL of extract at 100 bar and 55 °C for 2 h, achieving 10% inhibition with DPPH methods. The extract presented a potentially suitable impregnation of PLA for biomedical applications.

2.
Front Microbiol ; 14: 1210898, 2023.
Article in English | MEDLINE | ID: mdl-37383629

ABSTRACT

Introduction: The S-layer proteins are a class of self-assembling proteins that form bi-dimensional lattices named S-Layer on the cell surface of bacteria and archaea. The protein SlpA, which is the major constituent of the Lactobacillus acidophilus S-layer, contains in its C-terminus region (SlpA284 - 444), a protein domain (named here as SLAPTAG) responsible for the association of SlpA to the bacterial surface. SLAPTAG was adapted for the development of a novel affinity chromatography method: the SLAPTAG-based affinity chromatography (SAC). Methods: Proteins with different molecular weights or biochemical functions were fused in-frame to the SLAPTAG and efficiently purified by a Bacillus subtilis-derived affinity matrix (named Bio-Matrix or BM). Different binding and elution conditions were evaluated to establish an optimized protocol. Results: The binding equilibrium between SLAPTAG and BM was reached after a few minutes of incubation at 4°C, with an apparent dissociation constant (KD) of 4.3µM. A reporter protein (H6-GFP-SLAPTAG) was used to compare SAC protein purification efficiency against commercial immobilized metal affinity chromatography. No differences in protein purification performance were observed between the two methods. The stability and reusability of the BM were evaluated, and it was found that the matrix remained stable for more than a year. BM could be reused up to five times without a significant loss in performance. Additionally, the recovery of bound SLAP-tagged proteins was explored using proteolysis with a SLAP-tagged version of the HRV-3c protease (SLAPASE). This released the untagged GFP while the cut SLAPTAG and the SLAPASE were retained in the BM. As an alternative, iron nanoparticles were linked to the BM, resulting in BMmag. The BMmag was successfully adapted for a magnetic SAC, a technique with potential applications in high-throughput protein production and purification. Discussion: The SAC protocol can be adapted as a universal tool for the purification of recombinant proteins. Furthermore, the SAC protocol utilizes simple and low-cost reagents, making it suitable for in-house protein purification systems in laboratories worldwide. This enables the production of pure recombinant proteins for research, diagnosis, and the food industry.

3.
Front Microbiol ; 13: 1046640, 2022.
Article in English | MEDLINE | ID: mdl-36386645

ABSTRACT

Brucella spp. are the etiological agent of animal and human brucellosis. We have reported previously that cyclophilins of Brucella (CypA and CypB) are upregulated within the intraphagosomal replicative niche and required for stress adaptation and host intracellular survival and virulence. Here, we characterize B. abortus cyclophilins, CypA, and CypB from a biochemical standpoint by studying their PPIase activity, chaperone activity, and oligomer formation. Even though CypA and CypB are very similar in sequence and share identical chaperone and PPIase activities, we were able to identify outstanding differential features between them. A series of differential peptide loops were predicted when comparing CypA and CypB, differences that might explain why specific antibodies (anti-CypA or anti-CypB) were able to discriminate between both cyclophilins without cross-reactivity. In addition, we identified the presence of critical amino acids in CypB, such as the Trp134 which is responsible for the cyclosporin A inhibition, and the Cys128 that leads to CypB homodimer formation by establishing a disulfide bond. Here, we demonstrated that CypB dimer formation was fully required for stress adaptation, survival within HeLa cells, and mouse infection in B. abortus. The presence of Trp134 and the Cys128 in CypB, which are not present in CypA, suggested that two different kinds of cyclophilins have evolved in Brucella, one with eukaryotic features (CypB), another (CypA) with similar features to Gram-negative cyclophilins.

4.
Vaccine ; 40(8): 1065-1073, 2022 02 16.
Article in English | MEDLINE | ID: mdl-35086742

ABSTRACT

Shiga-toxin-producing Escherichia coli (STEC) is an important food-borne pathogen that causes hemorrhagic colitis and hemolytic uremic syndrome (HUS) in humans. Since no vaccines are available and antibiotic treatment is not recommended because promotes the appearance of HUS symptoms, the control of STEC intestinal colonization in cows, which is an important environmental reservoir, is crucial to control this zoonosis. Here, we evaluated the adaptation of an attenuated strain of Salmonella enterica serovar Typhimurium (ΔaroA mutant) as a vaccine platform for preventing STEC intestinal colonization that was studied in a mouse model. A chimeric antigen formed by the combination of the STEC peptides EspA36-192, Intimin653-935, Tir 258-361, and H7 flagellin352-374 (EITH7) was constructed and fused to the ß-lactamase signal sequence (bla SS) that drives the secretion of the chimeric antigen to the bacterial periplasmic space. Oral administration of ΔaroA-ST(EITH7) in a regime of three doses of immunization elicited both mucosal and humoral immune responses that protect mice against a STEC oral experimental infection. Remarkably, serum antibodies not only were able to bind the chimeric antigen EITH7 but also to block actin pedestal formation triggered by the type three secretion system (T3SS) in Enteropathogenic Escherichia coli (EPEC). Furthermore, a single-dose protocol was evaluated, and mice were orally immunized with ΔaroA-ST(EITH7). Interestingly, although with this protocol of immunization only fecal α-EITH7 IgA antibodies were induced and no α-EITH7 in sera were detected, mice were able to efficiently control an oral experimental infection with 1010 STEC (strain Escherichia coli O157:H7), suggesting that mucosal immune response was necessary and sufficient to control STEC intestinal colonization.


Subject(s)
Escherichia coli Infections , Escherichia coli Proteins , Escherichia coli Vaccines , Salmonella Vaccines , Shiga-Toxigenic Escherichia coli , Animals , Antibodies, Bacterial , Cattle , Escherichia coli Infections/prevention & control , Escherichia coli Proteins/genetics , Female , Mice , Salmonella typhimurium
5.
Front Microbiol ; 11: 509380, 2020.
Article in English | MEDLINE | ID: mdl-33193117

ABSTRACT

S-layers are bacterial structures present on the surface of several Gram-positive and Gram-negative bacteria that play a role in bacterial protection. In Lactobacillus acidophilus (L. acidophilus ATCC 4356), the S-layer is mainly composed of the protein SlpA. A tandem of two copies of the protein domain SLP-A (pfam: 03217) was identified at the C-terminal of SlpA, being this double SLP-A protein domain (in short dSLP-A) necessary and sufficient for the association of the protein to the L. acidophilus cell wall. A variety of proteins fused to the dSLP-A domain were able to spontaneously associate with high affinity to the cell wall of L. acidophilus and Bacillus subtilis var. natto, in a process that we termed decoration. Binding of dSLP-A-containing-proteins to L. acidophilus was stable at conditions that mimic the gastrointestinal transit in terms of pH, proteases, and bile salts. To evaluate if protein decoration of L. acidophilus can be adapted to generate an oral vaccine platform, a chimeric antigen derived from the bacterial pathogen Shiga-toxin-producing Escherichia coli (STEC) was constructed by fusing the sequences encoding the polypeptides EspA36-192, Intimin653-953, Tir240-378, and H7 flagellin352-374 (EITH7) to the dSLP-A domain (EITH7-dSLP-A). Recombinantly expressed EITH7-dSLP-A protein was affinity purified and combined with L. acidophilus cultures to allow the association of the chimeric antigen to the bacterial surface. EITH7-decorated L. acidophilus was orally administered to BALB/c mice and the induction of anti-EITH7 specific antibodies in sera and feces determined by ELISA. Mice presenting significantly higher anti-EITH7 antibodies titers were able to control more efficiently an experimental STEC infection than mice that received the non-decorated L. acidophilus carrier, indicating that antigen-decorated L. acidophilus can be adapted as a mucosal immunization delivery platform to elicit a protective immune response for vaccine purposes.

6.
Front Plant Sci ; 9: 1686, 2018.
Article in English | MEDLINE | ID: mdl-30515183

ABSTRACT

Mesorhizobium loti MAFF303099 is a rhizobial strain that nodulates Lotus spp. A M. loti MAFF303099 mutant strain affected in the tatC gene was generated. This strain presented an altered protein secretion level to the culture supernatant and also a higher sensitivity to SDS. Its nodulation phenotype on Lotus showed the induction of small and colorless nodules, and in a larger number than those induced by the wild-type strain. In addition, these nodules presented defects in the degree of occupation by rhizobia. Two Rieske Fe/S proteins, encoded by the mll2707 and mlr0970 genes, were predicted as potential Tat substrates in M. loti MAFF303099. The transcriptional expression of mll2707 and mlr0970 genes was analyzed under different oxygen growth conditions. The mll2707 gene was expressed constitutively, while the expression of the mlr0970 gene was only detected under anaerobic and microaerophilic in vitro conditions. Both genes were down-regulated in the tatC mutant strain. mll2707 and mlr0970 mRNAs from the wild-type strain were detected in nodules. Using a translational reporter peptide fusion, we found that the Mll2707 protein was only detectable in the wild-type strain. On the other hand, although Mlr0970 protein was detected in wild-type and tatC mutant strains, its association with the membrane was favored in the wild-type strain. The tatC and the mll2707 mutant strains were affected in the cytochrome c oxidase activity. These results confirm that Mll2707 is required for cytochrome c-dependent respiration and that Tat functionality is required for the correct activity of Mll2707. The mll2707 mutant strain showed a nodulation phenotype similar to the tatC mutant strain, although it presented only a slight difference in comparison with wild-type strain in terms of nodule occupation. No defective phenotype was observed in the nodulation with the mlr0970 mutant strain. These results indicate that, of the two Rieske Fe/S proteins coded by M. loti MAFF303099, only Mll2707 expression is required for the induction of effective nodules, and that the functionality of the Tat system is necessary not only for the correct function of this protein, but also for some other protein required in an earlier stage of the nodulation process.

7.
Medicina (B Aires) ; 77(3): 185-190, 2017.
Article in English | MEDLINE | ID: mdl-28643674

ABSTRACT

Shiga toxin (Stx)-producing Escherichia coli (STEC) infections are implicated in the development of the life-threatening hemolytic-uremic syndrome (HUS). Despite the magnitude of the social and economic problems caused by HUS, no licensed vaccine or effective therapy is currently available for human use. Prevention of STEC infections continues being the most important measure to reduce HUS incidence. This is especially true for Argentina where HUS incidence among children is extremely high and shows an endemic pattern. The aim of this work was to investigate serologically adult staff of kindergartens in Buenos Aires city and suburban areas in order to detect possible carriers, and to educate personnel about good practices to reduce HUS transmission. We also assessed the microbiological quality of water and meal samples from the same kindergartens. We tested 67 healthy adults, 13 water supplies and 6 meals belonging to 6 public kindergartens. We analysed hand swabs for isolation of STEC and serum samples for the presence of antibodies against Stx and lipopolysaccharide (LPS) of O157 serogroup. We identified 46 Stx2-positive individuals, but only 7 for O157 LPS. No presence of STEC pathogens was detected in hands of staff, water or meal samples.


Subject(s)
Escherichia coli Infections/prevention & control , Escherichia coli O157/isolation & purification , Hemolytic-Uremic Syndrome/microbiology , Hemolytic-Uremic Syndrome/prevention & control , Adult , Argentina/epidemiology , Child , Disease Outbreaks , Electrophoresis , Escherichia coli Infections/epidemiology , Escherichia coli Infections/microbiology , Escherichia coli Infections/transmission , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/epidemiology , Humans , Risk Factors , Serotyping , Urban Population
8.
Medicina (B.Aires) ; 77(3): 185-190, jun. 2017. graf
Article in English | LILACS | ID: biblio-894455

ABSTRACT

Shiga toxin (Stx)-producing Escherichia coli (STEC) infections are implicated in the development of the life-threatening hemolytic-uremic syndrome (HUS). Despite the magnitude of the social and economic problems caused by HUS, no licensed vaccine or effective therapy is currently available for human use. Prevention of STEC infections continues being the most important measure to reduce HUS incidence. This is especially true for Argentina where HUS incidence among children is extremely high and shows an endemic pattern. The aim of this work was to investigate serologically adult staff of kindergartens in Buenos Aires city and suburban areas in order to detect possible carriers, and to educate personnel about good practices to reduce HUS transmission. We also assessed the microbiological quality of water and meal samples from the same kindergartens. We tested 67 healthy adults, 13 water supplies and 6 meals belonging to 6 public kindergartens. We analysed hand swabs for isolation of STEC and serum samples for the presence of antibodies against Stx and lipopolysaccharide (LPS) of O157 serogroup. We identified 46 Stx2-positive individuals, but only 7 for O157 LPS. No presence of STEC pathogens was detected in hands of staff, water or meal samples.


Las infecciones bacterianas con Escherichia coli productor de toxina Shiga (Stx) (STEC) están implicadas en el desarrollo del síndrome urémico hemolítico (SUH). A pesar de la magnitud del problema social y económico causado por el SUH, actualmente no existe un tratamiento específico o una vacuna eficaz para uso humano. Por lo tanto, la prevención de las infecciones por STEC es la tarea central para reducir la incidencia del SUH. Esto es especialmente cierto para Argentina en donde el SUH muestra un comportamiento endémico y presenta una incidencia extremadamente alta entre los niños. En efecto, la mediana de casos notificados en menores de 5 años para el periodo 2010-2015 fue 306, mientras que la tasa de notificación fue 8.5 casos cada 100 000 menores/año (http://www.msal.gob.ar/images/stories/boletines/boletin_integrado_vigilancia_N335-SE45.pdf). El objetivo de este trabajo fue analizar serológicamente al personal adulto de jardines de infantes de la ciudad de Buenos Aires y el área suburbana con el fin de detectar portadores, y brindarles formación sobre las buenas prácticas para reducir la transmisión de infecciones con STEC y así evitar el SUH. También se evaluó la calidad microbiológica de las muestras de agua y de la comida elaborada en los mismos jardines. Hemos estudiado 67 adultos, a través del hisopado de manos para la búsqueda de STEC y suero para la presencia de anticuerpos contra Stx y el lipopolisacárido (LPS) de serogrupo O157. También se analizaron 13 suministros de agua y 6 muestras de comida pertenecientes a 6 jardines de infantes públicos. Se identificaron 46 individuos positivos para Stx2, pero solo 7 para LPS-O157. No se detectó presencia de patógenos STEC en las muestras de las manos del personal, ni en los reservorios de agua o muestras de comida.


Subject(s)
Humans , Child , Adult , Escherichia coli O157/isolation & purification , Escherichia coli Infections/prevention & control , Hemolytic-Uremic Syndrome/microbiology , Hemolytic-Uremic Syndrome/prevention & control , Argentina/epidemiology , Urban Population , Serotyping , Disease Outbreaks , Risk Factors , Electrophoresis , Escherichia coli Infections/microbiology , Escherichia coli Infections/transmission , Escherichia coli Infections/epidemiology , Hemolytic-Uremic Syndrome/blood
9.
Front Immunol ; 8: 171, 2017.
Article in English | MEDLINE | ID: mdl-28261222

ABSTRACT

Most pathogens infect through mucosal surfaces, and parenteral immunization typically fails to induce effective immune responses at these sites. Development of oral-administered vaccines capable of inducing mucosal as well as systemic immunity while bypassing the issues of antigen degradation and immune tolerance could be crucial for the control of enteropathogens. This study demonstrates that U-Omp19, a bacterial protease inhibitor with immunostimulatory features, coadministered with Salmonella antigens by the oral route, enhances mucosal and systemic immune responses in mice. U-Omp19 was able to increase antigen-specific production of IFN-γ and IL-17 and mucosal (IgA) antibody response. Finally, oral vaccination with U-Omp19 plus Salmonella antigens conferred protection against virulent challenge with Salmonella Typhimurium, with a significant reduction in bacterial loads. These findings prove the efficacy of this novel adjuvant in the Salmonella infection model and support the potential of U-Omp19 as a suitable adjuvant in oral vaccine formulations against mucosal pathogens requiring T helper (Th)1-Th17 protective immune responses.

10.
Cent European J Urol ; 69(3): 313, 2016.
Article in English | MEDLINE | ID: mdl-27730003
11.
J Control Release ; 220(Pt A): 18-28, 2015 Dec 28.
Article in English | MEDLINE | ID: mdl-26456256

ABSTRACT

We report here that a bacterial protease inhibitor from Brucella spp. called U-Omp19 behaves as an ideal constituent for a vaccine formulation against infectious diseases. When co-administered orally with an antigen (Ag), U-Omp19: i) can bypass the harsh environment of the gastrointestinal tract by inhibiting stomach and intestine proteases and consequently increases the half-life of the co-administered Ag at immune inductive sites: Peyer's patches and mesenteric lymph nodes while ii) it induces the recruitment and activation of antigen presenting cells (APCs) and increases the amount of intracellular Ag inside APCs. Therefore, mucosal as well as systemic Ag-specific immune responses, antibodies, Th1, Th17 and CD8(+) T cells are enhanced when U-Omp19 is co-administered with the Ag orally. Finally, this bacterial protease inhibitor in an oral vaccine formulation confers mucosal protection and reduces parasite loads after oral challenge with virulent Toxoplasma gondii.


Subject(s)
Antigens/metabolism , Bacterial Proteins/pharmacology , Brucella/chemistry , Immunity, Mucosal , Protease Inhibitors/pharmacology , Vaccines/immunology , Administration, Oral , Amino Acid Sequence , Animals , Female , Mice , Mice, Inbred Strains , Molecular Sequence Data
12.
Vaccine ; 33(19): 2248-2253, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25820069

ABSTRACT

Zoonoses that affect human and animal health have an important economic impact. In the study now presented, a bivalent vaccine has been developed that has the potential for preventing the transmission from cattle to humans of two bacterial pathogens: Brucella abortus and Shiga toxin-producing Escherichia coli (STEC). A 66kDa chimeric antigen, composed by EspA, Intimin, Tir, and H7 flagellin (EITH7) from STEC, was constructed and expressed in B. abortus Δpgm vaccine strain (BabΔpgm). Mice orally immunized with BabΔpgm(EITH7) elicited an immune response with the induction of anti-EITH7 antibodies (IgA) that clears an intestinal infection of E. coli O157:H7 three times faster (t=4 days) than mice immunized with BabΔpgm carrier strain (t=12 days). As expected, mice immunized with BabΔpgm(EITH7) strain also elicited a protective immune response against B. abortus infection. A Brucella-based vaccine platform is described capable of eliciting a combined protective immune response against two bacterial pathogens with diverse lifestyles-the intracellular pathogen B. abortus and the intestinal extracellular pathogen STEC.


Subject(s)
Bacterial Vaccines/immunology , Bacterial Vaccines/isolation & purification , Brucella abortus/immunology , Brucellosis, Bovine/prevention & control , Escherichia coli Infections/prevention & control , Escherichia coli O157/immunology , Animals , Antibodies, Bacterial/immunology , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Brucella abortus/genetics , Brucellosis, Bovine/immunology , Brucellosis, Bovine/microbiology , Cattle , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Escherichia coli O157/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/immunology , Gene Expression , Immunoglobulin A/immunology , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/isolation & purification
13.
PLoS One ; 9(7): e101279, 2014.
Article in English | MEDLINE | ID: mdl-24983999

ABSTRACT

Brucella, the etiological agent of animal and human brucellosis, is a bacterium with the capacity to modulate the inflammatory response. Cyclic ß-1,2-glucan (CßG) is a virulence factor key for the pathogenesis of Brucella as it is involved in the intracellular life cycle of the bacteria. Using comparative studies with different CßG mutants of Brucella, cgs (CßG synthase), cgt (CßG transporter) and cgm (CßG modifier), we have identified different roles for this polysaccharide in Brucella. While anionic CßG is required for bacterial growth in low osmolarity conditions, the sole requirement for a successful Brucella interaction with mammalian host is its transport to periplasmic space. Our results uncover a new role for CßG in promoting splenomegaly in mice. We showed that CßG-dependent spleen inflammation is the consequence of massive cell recruitment (monocytes, dendritics cells and neutrophils) due to the induction of pro-inflammatory cytokines such as IL-12 and TNF-α and also that the reduced splenomegaly response observed with the cgs mutant is not the consequence of changes in expression levels of the characterized Brucella PAMPs LPS, flagellin or OMP16/19. Complementation of cgs mutant with purified CßG increased significantly spleen inflammation response suggesting a direct role for this polysaccharide.


Subject(s)
Brucellosis/microbiology , Inflammation/microbiology , Splenomegaly/microbiology , beta-Glucans/metabolism , ATP-Binding Cassette Transporters/genetics , Animals , Brucella abortus/genetics , Brucella abortus/metabolism , Cytokines/metabolism , Gene Knockout Techniques , Glucosyltransferases/genetics , Mice
14.
Infect Immun ; 81(2): 521-30, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23230297

ABSTRACT

Brucella is an intracellular bacterial pathogen that causes the worldwide zoonotic disease brucellosis. Brucella virulence relies on its ability to transition to an intracellular lifestyle within host cells. Thus, this pathogen must sense its intracellular localization and then reprogram gene expression for survival within the host cell. A comparative proteomic investigation was performed to identify differentially expressed proteins potentially relevant for Brucella intracellular adaptation. Two proteins identified as cyclophilins (CypA and CypB) were overexpressed in the intracellular environment of the host cell in comparison to laboratory-grown Brucella. To define the potential role of cyclophilins in Brucella virulence, a double-deletion mutant was constructed and its resulting phenotype was characterized. The Brucella abortus ΔcypAB mutant displayed increased sensitivity to environmental stressors, such as oxidative stress, pH, and detergents. In addition, the B. abortus ΔcypAB mutant strain had a reduced growth rate at lower temperature, a phenotype associated with defective expression of cyclophilins in other microorganisms. The B. abortus ΔcypAB mutant also displays reduced virulence in BALB/c mice and defective intracellular survival in HeLa cells. These findings suggest that cyclophilins are important for Brucella virulence and survival in the host cells.


Subject(s)
Adaptation, Physiological/physiology , Brucella abortus/physiology , Brucellosis/microbiology , Cyclophilins/physiology , Stress, Physiological/physiology , Adaptation, Physiological/genetics , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Brucella abortus/genetics , Brucella abortus/metabolism , Brucella abortus/pathogenicity , Brucellosis/genetics , Brucellosis/metabolism , Cell Line, Tumor , Cyclophilins/genetics , Cyclophilins/metabolism , Female , Gene Expression/genetics , Gene Expression Regulation, Bacterial , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Mutation/genetics , Proteomics/methods , Stress, Physiological/genetics , Virulence
15.
Cell Host Microbe ; 6(3): 268-78, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19748468

ABSTRACT

The actin-based motility of the intracellular pathogen Listeria monocytogenes relies on ActA, a bacterial factor with a structural domain allowing it to mimic the actin nucleation-promoting activity of host cell proteins of the WASP/WAVE family. Here, we used an RNAi-based genetic approach in combination with computer-assisted image analysis to investigate the role of host factors in L. monocytogenes cell-to-cell spread. We showed that the host cell serine/threonine kinase CK2 is required for efficient actin tail formation by L. monocytogenes. Furthermore, CK2-mediated phosphorylation of ActA regulated its affinity for the actin-nucleating ARP2/3 complex, as is the case for CK2-mediated phosphorylation of WASP and WAVE. Thus, ActA not only displays structural mimicry of WASP/WAVE family members, but also regulatory mimicry, having precisely co-opted the host machinery regulating these proteins. Comparisons based on ActA amino acid sequence suggest that unrelated pathogens that display actin-based motility may have evolved a similar strategy of regulatory mimicry.


Subject(s)
Actin-Related Protein 2-3 Complex/metabolism , Bacterial Proteins/metabolism , Listeria monocytogenes/physiology , Listeriosis/metabolism , Membrane Proteins/metabolism , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Casein Kinase II/metabolism , Female , HeLa Cells , Humans , Listeria monocytogenes/chemistry , Listeria monocytogenes/genetics , Listeriosis/enzymology , Listeriosis/microbiology , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Protein Binding , Sequence Alignment
16.
Blood ; 111(3): 1404-12, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-17986662

ABSTRACT

Salmonella typhimurium engineered to deliver cancer/testis antigen NY-ESO-1 through type III secretion (S typhimurium-NY-ESO-1) was shown to be an efficient cancer vaccine construct in mice and to stimulate NY-ESO-1-specific CD8(+)/CD4(+) T cells in vitro in patients with cancer with NY-ESO-1 spontaneous immunity. We also showed that individuals without spontaneous immunity to NY-ESO-1 had specific CD4(+) T-cell precursors with high avidity to NY-ESO-1 under tight control by CD4(+)CD25(+) regulatory T (Treg) cells. We now found that in healthy donors and patients with melanoma without NY-ESO-1 spontaneous immunity, S typhimurium-NY-ESO-1 elicits CD4(+) T helper 1 (Th1) cells in vitro recognizing naturally processed antigen from these high-avidity NY-ESO-1-specific naive precursors. In contrast to peptide stimulation, induction of specific Th1 cells with S typhimurium-NY-ESO-1 did not require in vitro depletion of CD4(+)CD25(+) Treg cells, and this prevailing effect was partially blocked by disruption of interleukin-6 or glucocorticoid-induced TNF receptor (GITR) signals. Furthermore, S typhimurium-induced Th1 cells had higher GITR expression than peptide-induced Th1 cells and were resistant to suppression by CD4(+)CD25(+) Treg cells in a GITR-dependent fashion. We propose that S typhimurium-NY-ESO-1 induces antigen-specific T-cell responses that are resistant to suppression by CD4(+)CD25(+) Treg cells.


Subject(s)
Genetic Vectors/genetics , Salmonella typhimurium/genetics , Salmonella typhimurium/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Cell Differentiation/immunology , Cells, Cultured , Glucocorticoid-Induced TNFR-Related Protein , Humans , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-6/pharmacology , Lymphocyte Activation/immunology , Membrane Proteins/immunology , Membrane Proteins/metabolism , Receptors, Nerve Growth Factor/immunology , Receptors, Tumor Necrosis Factor/immunology , Sensitivity and Specificity , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/drug effects
17.
Infect Immun ; 74(10): 5826-33, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16988261

ABSTRACT

Type III protein secretion systems, which are organelles with the capacity to deliver bacterial proteins into host cells, have been adapted to deliver heterologous antigens for vaccine development. A limitation of these antigen delivery systems is that some proteins are not amenable to secretion through this pathway. We show here that proteins from the simian and human immunodeficiency viruses that are not permissive for secretion through a Salmonella enterica serovar Typhimurium type III secretion system can be modified to travel this secretion pathway by introduction of discrete mutations. Proteins optimized for secretion were presented more efficiently via the major histocompatibility complex class I pathway and were able to induce a better immune response.


Subject(s)
AIDS Vaccines/metabolism , Gene Products, gag/metabolism , HIV , Retroviridae Proteins/metabolism , Salmonella typhimurium/metabolism , Simian Immunodeficiency Virus , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Animals , Bacterial Proteins/metabolism , Female , Gene Products, gag/genetics , Gene Products, gag/immunology , HIV/immunology , Humans , Mice , Mice, Inbred BALB C , Mutation , Protein Transport , Retroviridae Proteins/genetics , Retroviridae Proteins/immunology , Salmonella typhimurium/genetics , Simian Immunodeficiency Virus/immunology
18.
J Clin Invest ; 116(7): 1946-54, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16794737

ABSTRACT

Bacterial vectors may offer many advantages over other antigen delivery systems for cancer vaccines. We engineered a Salmonella typhimurium vaccine strain to deliver the NY-ESO-1 tumor antigen (S. typhimurium-NY-ESO-1) through a type III protein secretion system. The S. typhimurium-NY-ESO-1 construct elicited NY-ESO-1-specific CD8+ and CD4+ T cells from peripheral blood lymphocytes of cancer patients in vitro. Oral administration of S. typhimurium-NY-ESO-1 to mice resulted in the regression of established NY-ESO-1-expressing tumors. Intratumoral inoculation of S. typhimurium-NY-ESO-1 to NY-ESO-1-negative tumors resulted in delivery of antigen in vivo and led to tumor regression in the presence of preexisting NY-ESO-1-specific CD8+ T cells. Specific T cell responses against at least 2 unrelated tumor antigens not contained in the vaccine were observed, demonstrating epitope spreading. We propose that antigen delivery through the S. typhimurium type III secretion system is a promising novel strategy for cancer vaccine development.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines , Membrane Proteins/immunology , Salmonella typhimurium/metabolism , Animals , Antibodies, Neoplasm/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/therapeutic use , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Epitopes , Female , Humans , Membrane Proteins/genetics , Membrane Proteins/therapeutic use , Mice , Mice, Inbred BALB C , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Salmonella typhimurium/genetics
19.
Glycobiology ; 16(7): 679-91, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16603625

ABSTRACT

Brucella abortus cyclic glucan synthase (Cgs) is a 320-kDa (2868-amino acid) polytopic integral inner membrane protein responsible for the synthesis of the virulence factor cyclic beta-1,2-glucan by a novel mechanism in which the enzyme itself acts as a protein intermediate. Cgs functions as an inverting processive beta-1,2-autoglucosyltransferase and has the three enzymatic activities required for the synthesis of the cyclic glucan: initiation, elongation, and cyclization. To gain further insight into the protein domains that are essential for the enzymatic activity, we have compared the Cgs sequence with other glycosyltransferases (GTs). This procedure allowed us to identify in the Cgs region (475-818) the widely spaced D, DxD, E/D, (Q/R)xxRW motif that is highly conserved in the active site of numerous GTs. By site-directed mutagenesis and in vitro and in vivo activity assays, we have demonstrated that most of the amino acid residues of this motif are essential for Cgs activity. These sequence and site-directed mutagenesis analyses also indicate that Cgs should be considered a bi-functional modular GT, with an N-terminal GT domain belonging to a new GT family related to GT-2 (GT-84) followed by a GH-94 glycoside hydrolase C-terminal domain. Furthermore, over-expression of inactive mutants results in wild-type (WT) production of cyclic glucan when bacteria co-express the mutant and the WT form, indicating that Cgs may function in the membrane as a monomeric enzyme. Together, these results are compatible with a single addition model by which Cgs acts in the membrane as a monomer and uses the identified motif to form a single center for substrate binding and glycosyl-transfer reaction.


Subject(s)
Bacterial Proteins/chemistry , Brucella abortus/enzymology , Glycosyltransferases/chemistry , Virulence Factors/chemistry , beta-Glucans/metabolism , Amino Acid Sequence , Amino Acid Substitution , Bacterial Proteins/genetics , Binding Sites/genetics , Brucella abortus/pathogenicity , Conserved Sequence , Glycosyltransferases/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Protein Structure, Tertiary , Sequence Alignment , Sequence Analysis, Protein , Virulence Factors/genetics
20.
Infect Immun ; 74(2): 1084-90, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16428755

ABSTRACT

Central to the study of type III secretion systems is the availability of reporter systems to monitor bacterial protein translocation into host cells. We report here the development of a bacteriophage P1 Cre recombinase-based system to monitor the translocation of bacterial proteins into mammalian cells. Bacteriophage P1 Cre recombinase fused to the secretion and translocation signals of Salmonella enterica serovar Typhimurium of the type III secreted protein SopE was secreted in a type III secretion system-dependent fashion. More importantly, the SopE-Cre chimera was translocated into host cells via the type III secretion system and activated the expression of luciferase and green fluorescent protein reporters of Cre recombinase activity.


Subject(s)
Bacterial Proteins/metabolism , Bacteriophage P1/enzymology , Genes, Reporter , Integrases/metabolism , Recombinant Fusion Proteins/metabolism , Salmonella typhimurium/pathogenicity , Viral Proteins/metabolism , Animals , Bacterial Proteins/genetics , Bacteriophage P1/genetics , COS Cells , Chlorocebus aethiops , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Integrases/genetics , Luciferases/genetics , Luciferases/metabolism , Mice , Plasmids , Protein Transport , Recombinant Fusion Proteins/genetics , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...