Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 180
Filtrar
Más filtros

Medicinas Complementárias
Métodos Terapéuticos y Terapias MTCI
Intervalo de año de publicación
1.
Cells ; 10(10)2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34685593

RESUMEN

TLR5 ligand flagellin-containing fusion proteins are potential vaccine candidates for many diseases. A recombinant fusion protein of flagellin A and the major birch pollen allergen Bet v 1 (rFlaA:Betv1) modulates immune responses in vitro and in vivo. We studied the effects of rFlaA:Betv1 on bone marrow-derived macrophages (BMDMs). BMDMs differentiated from BALB/c, C57BL/6, TLR5-/-, or MyD88-/- mice were pre-treated with inhibitors, stimulated with rFlaA:Betv1 or respective controls, and analyzed for activation, cytokine secretion, metabolic state, RNA transcriptome, and modulation of allergen-specific Th2 responses. Stimulation of BMDMs with rFlaA:Betv1 resulted in MyD88-dependent production of IL-1ß, IL-6, TNF-α, IL-10, CD69 upregulation, and a pronounced shift towards glycolysis paralleled by activation of MAPK, NFκB, and mTOR signaling. Inhibition of either mTOR (rapamycin) or SAP/JNK-MAPK signaling (SP600125) resulted in dose-dependent metabolic suppression. In BMDM and T cell co-cultures, rFlaA:Betv1 stimulation suppressed rBet v 1-induced IL-5 and IL-13 secretion while inducing IFN-γ production. mRNA-Seq analyses showed HIF-1a, JAK, STAT, phagosome, NLR, NFκB, TNF, TLR, and chemokine signaling to participate in the interplay of cell activation, glycolysis, and immune response. rFlaA:Betv1 strongly activated BMDMs, resulting in MyD88-, MAPK-, and mTOR-dependent enhancement of glucose metabolism. Our results suggest macrophages are important target cells to consider during restauration of allergen tolerance during AIT.


Asunto(s)
Alérgenos/inmunología , Antígenos Bacterianos/inmunología , Antígenos de Plantas/inmunología , Flagelina/inmunología , Proteínas Recombinantes de Fusión/inmunología , Animales , Proteínas Bacterianas/inmunología , Células Cultivadas , Glucosa/metabolismo , Macrófagos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Plantas/inmunología , Polen/inmunología
2.
PLoS Pathog ; 17(9): e1009930, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34496007

RESUMEN

Fatty acid-derived acyl chains of phospholipids and lipoproteins are central to bacterial membrane fluidity and lipoprotein function. Though it can incorporate exogenous unsaturated fatty acids (UFA), Staphylococcus aureus synthesizes branched chain fatty acids (BCFA), not UFA, to modulate or increase membrane fluidity. However, both endogenous BCFA and exogenous UFA can be attached to bacterial lipoproteins. Furthermore, S. aureus membrane lipid content varies based upon the amount of exogenous lipid in the environment. Thus far, the relevance of acyl chain diversity within the S. aureus cell envelope is limited to the observation that attachment of UFA to lipoproteins enhances cytokine secretion by cell lines in a TLR2-dependent manner. Here, we leveraged a BCFA auxotroph of S. aureus and determined that driving UFA incorporation disrupted infection dynamics and increased cytokine production in the liver during systemic infection of mice. In contrast, infection of TLR2-deficient mice restored inflammatory cytokines and bacterial burden to wildtype levels, linking the shift in acyl chain composition toward UFA to detrimental immune activation in vivo. In in vitro studies, bacterial lipoproteins isolated from UFA-supplemented cultures were resistant to lipase-mediated ester hydrolysis and exhibited heightened TLR2-dependent innate cell activation, whereas lipoproteins with BCFA esters were completely inactivated after lipase treatment. These results suggest that de novo synthesis of BCFA reduces lipoprotein-mediated TLR2 activation and improves lipase-mediated hydrolysis making it an important determinant of innate immunity. Overall, this study highlights the potential relevance of cell envelope acyl chain repertoire in infection dynamics of bacterial pathogens.


Asunto(s)
Ácidos Grasos/inmunología , Ácidos Grasos/metabolismo , Inmunidad Innata/inmunología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/metabolismo , Animales , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Fluidez de la Membrana/fisiología , Ratones , Staphylococcus aureus/inmunología , Staphylococcus aureus/metabolismo
4.
EBioMedicine ; 59: 102867, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32651162

RESUMEN

BACKGROUND: Chronic and recurrent bacterial diseases are recalcitrant to treatment due to the ability of the causative agents to establish biofilms, thus development of means to prevent or resolve these structures are greatly needed. Our approach targets the DNABII family of bacterial DNA-binding proteins, which serve as critical structural components within the extracellular DNA scaffold of biofilms formed by all bacterial species tested to date. DNABII-directed antibodies rapidly disrupt biofilms and release the resident bacteria which promote their subsequent clearance by either host immune effectors or antibiotics that are now effective at a notably reduced concentration. METHODS: First, as a therapeutic approach, we used intact IgG or Fab fragments against a chimeric peptide immunogen designed to target protective epitopes within the DNA-binding tip domains of integration host factor to disrupt established biofilms in vitro and to mediate resolution of existing disease in vivo. Second, we performed preventative active immunisation with the chimeric peptide to induce the formation of antibody that blocks biofilm formation and disease development in a model of viral-bacterial superinfection. Further, toward the path for clinical use, we humanised a monoclonal antibody against the chimeric peptide immunogen, then characterised and validated that it maintained therapeutic efficacy. FINDINGS: We demonstrated efficacy of each approach in two well-established pre-clinical models of otitis media induced by the prevalent respiratory tract pathogen nontypeable Haemophilus influenzae, a common biofilm disease. INTERPRETATION: Collectively, our data revealed two approaches with substantive efficacy and potential for broad application to combat diseases with a biofilm component. FUNDING: Supported by R01 DC011818 to LOB and SDG.


Asunto(s)
Antibacterianos/farmacología , Anticuerpos Monoclonales/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Biopelículas/efectos de los fármacos , AdnB Helicasas/antagonistas & inhibidores , Péptidos/farmacología , Animales , Anticuerpos Monoclonales/inmunología , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/microbiología , Infecciones Bacterianas/prevención & control , Proteínas Bacterianas/inmunología , AdnB Helicasas/inmunología , Evaluación Preclínica de Medicamentos , Humanos , Ratones , Péptidos/química , Péptidos/inmunología , Conejos
5.
Int J Nanomedicine ; 15: 3877-3886, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32581535

RESUMEN

INTRODUCTION: Vaccine formulation with appropriate adjuvants is an attractive approach to develop protective immunity against pathogens. Calcium phosphate nanoparticles (CaPNs) are considered as ideal adjuvants and delivery systems because of their great potential for enhancing immune responses. In the current study, we have designed nanoparticle-based vaccine candidates to induce immune responses and protection against B. melitensis and B. abortus. MATERIALS AND METHODS: For this purpose, we used three Brucella antigens (FliC, 7α-HSDH, BhuA) and two multi-epitopes (poly B and poly T) absorbed by CaPNs. The efficacy of each formulation was evaluated by measuring humoral, cellular and protective responses in immunized mice. RESULTS: The CaPNs showed an average size of about 90 nm with spherical shape and smooth surface. The CaPNs-adsorbed proteins displayed significant increase in cellular and humoral immune responses compared to the control groups. In addition, our results showed increased ratio of specific IgG2a (associated with Th1) to specific IgG1 (associated with Th2). Also, immunized mice with different vaccine candidate formulations were protected against B. melitensis 16M and B. abortus 544, and showed same levels of protection as commercial vaccines (B. melitensis Rev.1 and B. abortus RB51) except for BhuA-CaPNs. DISCUSSION: Our data support the hypothesis that these antigens absorbed with CaPNs could be effective vaccine candidates against B. melitensis and B. abortus.


Asunto(s)
Antígenos Bacterianos/química , Vacuna contra la Brucelosis/química , Vacuna contra la Brucelosis/inmunología , Nanopartículas/química , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/farmacología , Animales , Proteínas Bacterianas/inmunología , Brucella abortus/inmunología , Brucella melitensis/inmunología , Brucelosis/inmunología , Brucelosis/prevención & control , Fosfatos de Calcio/química , Sistemas de Liberación de Medicamentos , Femenino , Inmunidad Humoral , Inmunización , Inmunoglobulina G/inmunología , Proteínas de Transporte de Membrana/inmunología , Ratones Endogámicos BALB C
6.
Int J Biol Macromol ; 161: 875-890, 2020 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-32535205

RESUMEN

This study reports a ≅12.5 kDa protein tetrachloro-1,4-benzoquinone reductase (CpsD) from Bacillus cereus strain AOA-CPS1 (BcAOA). CpsD is purified to homogeneity with a total yield of 35% and specific activity of 160 U·mg-1 of protein. CpsD showed optimal activity at pH 7.5 and 40 °C. The enzyme was found to be functionally stable between pH 7.0-7.5 and temperature between 30 °C and 35 °C. CpsD activity was enhanced by Fe2+ and inhibited by sodium azide and SDS. CpsD followed Michaelis-Menten kinetic exhibiting an apparent vmax, Km, kcat and kcat/Km values of 0.071 µmol·s-1, 94 µmol, 0.029 s-1 and 3.13 × 10-4 s-1·µmol-1, respectively, for substrate tetrachloro-1,4-benzoquinone. The bioinformatics analysis indicated that CpsD belongs to the PCD/DCoH superfamily, with specific conserved protein domains of pterin-4α-carbinolamine  dehydratase (PCD). This study proposed that CpsD catalysed the reduction of tetrachloro-1,4-benzoquinone to tetrachloro-p-hydroquinone and released the products found in phenylalanine hydroxylation system (PheOHS) via a Ping-Pong or atypical ternary mechanism; and regulate expression of phenylalanine 4-monooxygenase by blocking reverse flux in BcAOA PheOHS using a probable Yin-Yang mechanism. The study also concluded that CpsD may play a catalytic and regulatory role in BcAOA PheOHS and pentachlorophenol degradation pathway.


Asunto(s)
Bacillus cereus/metabolismo , Proteínas Bacterianas/inmunología , Cloranilo/metabolismo , Galactosiltransferasas/inmunología , Hidroxilación/fisiología , Pentaclorofenol/metabolismo , Fenilalanina/metabolismo , Cinética , Oxidorreductasas/metabolismo
7.
Infect Immun ; 88(8)2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32393506

RESUMEN

Bacillus anthracis is the causative agent of anthrax disease, presents with high mortality, and has been at the center of bioweapon efforts. The only currently U.S. FDA-approved vaccine to prevent anthrax in humans is anthrax vaccine adsorbed (AVA), which is protective in several animal models and induces neutralizing antibodies against protective antigen (PA), the cell-binding component of anthrax toxin. However, AVA requires a five-course regimen to induce immunity, along with an annual booster, and is composed of undefined culture supernatants from a PA-secreting strain. In addition, it appears to be ineffective against strains that lack anthrax toxin. Here, we investigated a vaccine formulation consisting of recombinant proteins from a surface-localized heme transport system containing near-iron transporter (NEAT) domains and its efficacy as a vaccine for anthrax disease. The cocktail of five NEAT domains was protective against a lethal challenge of inhaled bacillus spores at 3 and 28 weeks after vaccination. The reduction of the formulation to three NEATs (IsdX1, IsdX2, and Bslk) was as effective as a five-NEAT domain cocktail. The adjuvant alum, approved for use in humans, was as protective as Freund's Adjuvant, and protective vaccination correlated with increased anti-NEAT antibody reactivity and reduced bacterial levels in organs. Finally, the passive transfer of anti-NEAT antisera reduced mortality and disease severity, suggesting the protective component is comprised of antibodies. Collectively, these results provide evidence that a vaccine based upon recombinant NEAT proteins should be considered in the development of a next-generation anthrax vaccine.


Asunto(s)
Vacunas contra el Carbunco/inmunología , Carbunco/prevención & control , Anticuerpos Antibacterianos/biosíntesis , Anticuerpos Neutralizantes/biosíntesis , Antígenos Bacterianos/inmunología , Bacillus anthracis/efectos de los fármacos , Administración por Inhalación , Compuestos de Alumbre/administración & dosificación , Animales , Carbunco/inmunología , Carbunco/microbiología , Carbunco/mortalidad , Vacunas contra el Carbunco/administración & dosificación , Vacunas contra el Carbunco/genética , Antígenos Bacterianos/administración & dosificación , Antígenos Bacterianos/genética , Bacillus anthracis/inmunología , Bacillus anthracis/patogenicidad , Proteínas Bacterianas/administración & dosificación , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Proteínas Portadoras/administración & dosificación , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Complemento C5/deficiencia , Femenino , Adyuvante de Freund/administración & dosificación , Humanos , Inmunogenicidad Vacunal , Ratones Noqueados , Análisis de Supervivencia , Vacunación/métodos
8.
Front Immunol ; 10: 1349, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31293568

RESUMEN

Liposomes have been long considered as a vaccine delivery system but this technology remains to be fully utilized. Here, we describe a novel liposome-based subunit vaccine formulation for tuberculosis (TB) based on phosphatidylserine encapsulating two prominent TB antigens, Ag85B, and ESAT-6. We show that the resulting liposomes (Lipo-AE) are stable upon storage and can be readily taken up by antigen presenting cells and that their antigenic cargo is delivered and processed within endosomal cell compartments. The Lipo-AE vaccine formulation combined with the PolyIC adjuvant induced a mixed Th1/Th17-Th2 immune response to Ag85B but only a weak response to ESAT-6. An immunization regimen based on systemic delivery followed by mucosal boost with Lipo-AE resulted in the accumulation of resident memory T cells in the lungs. Most importantly though, when Lipo-AE vaccine candidate was administered to BCG-immunized mice subsequently challenged with low dose aerosol Mycobacterium tuberculosis, we observed a significant reduction of the bacterial load in the lungs and spleen compared to BCG alone. We therefore conclude that the immunization with mycobacterial antigens delivered by phosphatidylserine based liposomes in combination with Poly:IC adjuvant may represent a novel BCG boosting vaccination strategy.


Asunto(s)
Aciltransferasas/inmunología , Antígenos Bacterianos/inmunología , Vacuna BCG/inmunología , Proteínas Bacterianas/inmunología , Liposomas/inmunología , Tuberculosis Pulmonar/prevención & control , Adyuvantes Inmunológicos/administración & dosificación , Animales , Carga Bacteriana , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Memoria Inmunológica/inmunología , Pulmón/microbiología , Ratones , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/inmunología , Fosfatidilserinas/inmunología , Poli I-C/inmunología , Bazo/microbiología , Linfocitos T Colaboradores-Inductores/inmunología , Vacunación , Vacunas de Subunidad/inmunología
9.
PLoS Pathog ; 15(6): e1007877, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31226163

RESUMEN

Rapid bone destruction often leads to permanent joint dysfunction in patients with septic arthritis, which is mainly caused by Staphylococcus aureus (S. aureus). Staphylococcal cell wall components are known to induce joint inflammation and bone destruction. Here, we show that a single intra-articular injection of S. aureus lipoproteins (Lpps) into mouse knee joints induced chronic destructive macroscopic arthritis through TLR2. Arthritis was characterized by rapid infiltration of neutrophils and monocytes. The arthritogenic effect was mediated mainly by macrophages/monocytes and partially via TNF-α but not by neutrophils. Surprisingly, a S. aureus mutant lacking Lpp diacylglyceryl transferase (lgt) caused more severe joint inflammation, which coincided with higher bacterial loads of the lgt mutant in local joints than those of its parental strain. Coinjection of pathogenic S. aureus LS-1 with staphylococcal Lpps into mouse knee joints caused improved bacterial elimination and diminished bone erosion. The protective effect of the Lpps was mediated by their lipid moiety and was fully dependent on TLR2 and neutrophils. The blocking of CXCR2 on neutrophils resulted in total abrogation of the protective effect of the Lpps. Our data demonstrate that S. aureus Lpps elicit innate immune responses, resulting in a double-edged effect. On the one hand, staphylococcal Lpps boost septic arthritis. On the other hand, Lpps act as adjuvants and activate innate immunity, which could be useful for combating infections with multiple drug-resistant strains.


Asunto(s)
Artritis/inmunología , Proteínas Bacterianas/inmunología , Lipoproteínas/inmunología , Neutrófilos/inmunología , Staphylococcus aureus/inmunología , Animales , Artritis/genética , Artritis/microbiología , Artritis/patología , Proteínas Bacterianas/genética , Femenino , Lipoproteínas/genética , Ratones , Neutrófilos/patología , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/inmunología , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidad , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
10.
mBio ; 9(5)2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30327445

RESUMEN

Bacillus anthracis and Yersinia pestis, the causative agents of anthrax and plague, respectively, are two of the deadliest pathogenic bacteria that have been used as biological warfare agents. Although Biothrax is a licensed vaccine against anthrax, no Food and Drug Administration-approved vaccine exists for plague. Here, we report the development of a dual anthrax-plague nanoparticle vaccine employing bacteriophage (phage) T4 as a platform. Using an in vitro assembly system, the 120- by 86-nm heads (capsids) of phage T4 were arrayed with anthrax and plague antigens fused to the small outer capsid protein Soc (9 kDa). The antigens included the anthrax protective antigen (PA) (83 kDa) and the mutated (mut) capsular antigen F1 and the low-calcium-response V antigen of the type 3 secretion system from Y. pestis (F1mutV) (56 kDa). These viral nanoparticles elicited robust anthrax- and plague-specific immune responses and provided complete protection against inhalational anthrax and/or pneumonic plague in three animal challenge models, namely, mice, rats, and rabbits. Protection was demonstrated even when the animals were simultaneously challenged with lethal doses of both anthrax lethal toxin and Y. pestis CO92 bacteria. Unlike the traditional subunit vaccines, the phage T4 vaccine uses a highly stable nanoparticle scaffold, provides multivalency, requires no adjuvant, and elicits broad T-helper 1 and 2 immune responses that are essential for complete clearance of bacteria during infection. Therefore, phage T4 is a unique nanoparticle platform to formulate multivalent vaccines against high-risk pathogens for national preparedness against potential bioterror attacks and emerging infections.IMPORTANCE Following the deadly anthrax attacks of 2001, the Centers for Disease Control and Prevention (CDC) determined that Bacillus anthracis and Yersinia pestis that cause anthrax and plague, respectively, are two Tier 1 select agents that pose the greatest threat to the national security of the United States. Both cause rapid death, in 3 to 6 days, of exposed individuals. We engineered a virus nanoparticle vaccine using bacteriophage T4 by incorporating key antigens of both B. anthracis and Y. pestis into one formulation. Two doses of this vaccine provided complete protection against both inhalational anthrax and pneumonic plague in animal models. This dual anthrax-plague vaccine is a strong candidate for stockpiling against a potential bioterror attack involving either one or both of these biothreat agents. Further, our results establish the T4 nanoparticle as a novel platform to develop multivalent vaccines against pathogens of high public health significance.


Asunto(s)
Vacunas contra el Carbunco/inmunología , Carbunco/prevención & control , Antígenos Bacterianos/inmunología , Bacteriófago T4 , Vacuna contra la Peste/inmunología , Peste/prevención & control , Infecciones del Sistema Respiratorio/prevención & control , Animales , Anticuerpos Antibacterianos/sangre , Bacillus anthracis , Proteínas Bacterianas/inmunología , Toxinas Bacterianas/inmunología , Proteínas de la Cápside/inmunología , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Nanopartículas , Proteínas Citotóxicas Formadoras de Poros/inmunología , Conejos , Ratas , Células TH1/inmunología , Células Th2/inmunología , Yersinia pestis
11.
Vaccine ; 36(40): 6017-6021, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30172633

RESUMEN

Clostridium difficile flagellin FliC is a highly immunogenic pathogen-associated molecular pattern playing a key role in C. difficile pathogenesis and gut colonization. Here, we designed an oral vaccine against C. difficile with FliC encapsulated into pectin beads for colonic release. Bead stability and FliC retention was confirmed in vitro using simulated intestinal media (SIM), while bead degradation and FliC release was observed upon incubation in simulated colonic media (SCM). The importance of FliC encapsulation into pectin beads for protection against C. difficile was assessed in a vaccination assay using a lethal hamster model of C. difficile infection. Three groups of hamsters orally received either FliC-loaded beads or unloaded beads in gastro-resistant capsule to limit gastric degradation or free FliC. Two other groups were immunized with free FliC, one intra-rectally and the other intra-peritoneally. Hamsters were then challenged with a lethal dose of C. difficile VPI 10463. Fifty percent of hamsters orally immunized with FliC-loaded beads survived whereas all hamsters orally immunized with free FliC died within 7 days post challenge. No significant protection was observed in the other groups. Only intra-peritoneally immunized hamsters presented anti-FliC IgG antibodies in sera after immunizations. These results suggest that an oral immunization with FliC-loaded beads probably induced a mucosal immune response, therefore providing a protective effect. This study confirms the importance of FliC encapsulation into pectin beads for a protective oral vaccine against C. difficile.


Asunto(s)
Vacunas Bacterianas/inmunología , Infecciones por Clostridium/prevención & control , Flagelina/inmunología , Inmunidad Mucosa , Pectinas/administración & dosificación , Administración Oral , Animales , Proteínas Bacterianas/inmunología , Vacunas Bacterianas/química , Cápsulas , Clostridioides difficile , Colon/inmunología , Colon/microbiología , Cricetinae , Modelos Animales de Enfermedad , Femenino , Inmunoglobulina G/sangre , Microesferas , Vacunación/métodos
12.
Infect Immun ; 86(10)2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30104212

RESUMEN

Novel adjuvants are in demand for improving the efficacy of human vaccines. The immunomodulatory properties of Mycobacterium tuberculosis cell wall components have been highlighted in the formulation of complete Freund's adjuvant (CFA). We have explored the adjuvant potential of poly-α-l-glutamine (PLG), a lesser-known constituent of the pathogenic mycobacterial cell wall. Immune parameters indicated that the adjuvant potency of PLG was statistically comparable to that of CFA and better than that of alum in the context of H1 antigen (Ag85B and ESAT-6 fusion). At 1 mg/dose, PLG augmented the immune response of Ag85B, BP26, and protective antigen (PA) by increasing serum antibodies and cytokines in the culture supernatant of antigen-stimulated splenocytes. PLG modulated the humoral response of vaccine candidate ESAT-6, eliciting significantly higher levels of total IgG and isotypes (IgG1, IgG2a, and IgG2b). Additionally, the splenocytes from PLG-adjuvanted mice displayed a robust increase in the Th1-specific gamma interferon, tumor necrosis factor alpha, interleukin-2 (IL-2), Th2-specific IL-6 and IL-10, and Th17-specific IL-17A cytokines upon antigenic stimulation. PLG improved the protective efficacy of ESAT-6 by reducing bacillary load in the lung and spleen as well as granuloma formation, and it helped in maintaining vital health parameters of mice challenged with M. tuberculosis The median survival time of PLG-adjuvanted mice was 205 days, compared to 146 days for dimethyl-dioctadecyl ammonium bromide-monophosphoryl lipid A (DDA-MPL)-vaccinated groups and 224 days for Mycobacterium bovis BCG-vaccinated groups. PLG enhanced the efficiency of the ESAT-6 vaccine to the level of BCG and better than that of DDA-MPL (P < 0.05), with no ill effect in C57BL/6J mice. Our results propose that PLG is a promising adjuvant candidate for advanced experimentation.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Pared Celular/inmunología , Mycobacterium tuberculosis/inmunología , Péptidos/inmunología , Tuberculosis/microbiología , Aciltransferasas/administración & dosificación , Aciltransferasas/genética , Aciltransferasas/inmunología , Animales , Anticuerpos Antibacterianos , Antígenos Bacterianos/administración & dosificación , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/administración & dosificación , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Pared Celular/genética , Femenino , Adyuvante de Freund/inmunología , Humanos , Interleucina-17/genética , Interleucina-17/inmunología , Interleucina-2/genética , Interleucina-2/inmunología , Ratones , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/genética , Células TH1/inmunología , Tuberculosis/genética , Tuberculosis/inmunología , Tuberculosis/prevención & control , Vacunas contra la Tuberculosis/administración & dosificación , Vacunas contra la Tuberculosis/genética , Vacunas contra la Tuberculosis/inmunología
13.
Microb Pathog ; 124: 250-257, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30149131

RESUMEN

Brucellosis is an infectious disease that brings enormous economic burdens for developing countries. The Brucella melitensis (B. melitensis) M5-90 vaccine strain (M5-90) has been used on a large scale in China, but may cause abortions if given to pregnant goats or sheep subcutaneously during the late stages of gestation. Moreover, the vaccine M5-90 cannot differentiate natural from vaccinated infection. Therefore, a safer and more potent M5-90 vaccine is required. In this study, a vjbR mutant of M5-90 (M5-90ΔvjbR) was constructed and overcame these drawbacks. M5-90ΔvjbR strain showed reduced survival capability in murine macrophages (RAW 264.7) and BALB/c mice and induced high protective immunity in mice. In addition, M5-90ΔvjbR induced an anti-Brucella-specific immunoglobulin G (IgG) response and stimulated the expression of gamma interferon (INF-γ) and interleukin-4 (IL-4) in vaccinated mice. Furthermore, M5-90ΔvjbR induced IgG response and stimulated the secretion of IFN-γ and IL-4 in immunized sheep. Moreover, the VjbR antigen allowed serological differentiation between infected and vaccinated animals. These results suggest that M5-90ΔvjbR is an ideal live attenuated and efficacious live vaccine candidate against B. melitensis 16 M infection.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Vacuna contra la Brucelosis/inmunología , Brucella melitensis/inmunología , Brucelosis/prevención & control , Modelos Animales de Enfermedad , Animales , Anticuerpos Antibacterianos/inmunología , Proteínas Bacterianas/administración & dosificación , Vacuna contra la Brucelosis/administración & dosificación , Vacuna contra la Brucelosis/genética , Brucella melitensis/genética , Brucelosis/inmunología , Brucelosis/microbiología , Evaluación Preclínica de Medicamentos , Femenino , Eliminación de Gen , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Ratones , Ratones Endogámicos BALB C , Eliminación de Secuencia , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología
14.
Artículo en Inglés | MEDLINE | ID: mdl-30017079

RESUMEN

Streptococcus suis is a major Gram-positive swine pathogen associated with a wide variety of diseases in pigs. The efforts made to develop vaccines against this pathogen have failed because of lack of common cross-reactive antigens against different serotypes. Nowadays the interest has moved to surface and secreted proteins, as they have the highest chances to raise an effective immune response because they are in direct contact with host cells and are really exposed and accessible to antibodies. In this work, we have performed a comparative immunosecretomic approach to identify a set of immunoreactive secreted proteins common to the most prevalent serotypes of S. suis. Among the 67 proteins identified, three (SSU0020, SSU0934, and SSU0215) were those predicted extracellular proteins most widely found within the studied serotypes. These immunoreactive proteins may be interesting targets for future vaccine development as they could provide possible cross-reactivity among different serotypes of this pathogen.


Asunto(s)
Antígenos Bacterianos/aislamiento & purificación , Proteínas Bacterianas/aislamiento & purificación , Infecciones Estreptocócicas/veterinaria , Streptococcus suis/inmunología , Enfermedades de los Porcinos/prevención & control , Animales , Antígenos Bacterianos/biosíntesis , Antígenos Bacterianos/inmunología , Artritis/inmunología , Artritis/microbiología , Artritis/prevención & control , Artritis/veterinaria , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Bronconeumonía/inmunología , Bronconeumonía/microbiología , Bronconeumonía/prevención & control , Bronconeumonía/veterinaria , Electroforesis en Gel Bidimensional , Meningitis/inmunología , Meningitis/microbiología , Meningitis/prevención & control , Meningitis/veterinaria , Serogrupo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/prevención & control , Vacunas Estreptocócicas/biosíntesis , Streptococcus suis/crecimiento & desarrollo , Streptococcus suis/metabolismo , Streptococcus suis/patogenicidad , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/microbiología
15.
PLoS One ; 13(6): e0198207, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29856812

RESUMEN

GALT is an important antigen of Actinobacillus pleuropneumoniae (APP), which was shown to provide partial protection against APP infection in a previous study in our lab. The main purpose of the present study is to investigate GALT induced cross-protection between different APP serotypes and elucidate key mechanisms of the immune response to GALT antigenic stimulation. Bioinformatic analysis demonstrated that galT is a highly conserved gene in APP, widely distributed across multiple pathogenic strains. Homologies between any two strains ranges from 78.9% to 100% regarding the galT locus. Indirect enzyme-linked immunosorbent assay (ELISA) confirmed that GALT specific antibodies could not be induced by inactivated APP L20 or MS71 whole cell bacterin preparations. A recombinant fusion GALT protein derived from APP L20, however has proven to be an effective cross-protective antigen against APP sevorar 1 MS71 (50%, 4/8) and APP sevorar 5b L20 (75%, 6/8). Histopathological examinations have confirmed that recombinant GALT vaccinated animals showed less severe pathological signs in lung tissues than negative controls after APP challenge. Immunohistochemical (IHC) analysis indicated that the infiltration of neutrophils in the negative group is significantly increased compared with that in the normal control (P<0.001) and that in surviving animals is decreased compared to the negative group. Anti-GALT antibodies were shown to mediate phagocytosis of neutrophils. After interaction with anti-GALT antibodies, survival rate of APP challenged vaccinated animals was significantly reduced (P<0.001). This study demonstrated that GALT is an effective cross-protective antigen, which could be used as a potential vaccine candidate against multiple APP serotypes.


Asunto(s)
Infecciones por Actinobacillus/veterinaria , Actinobacillus pleuropneumoniae/inmunología , Antígenos Bacterianos/inmunología , Vacunas Bacterianas/inmunología , Pleuroneumonía/veterinaria , Enfermedades de los Porcinos/prevención & control , UDP-Glucosa-Hexosa-1-Fosfato Uridiltransferasa/inmunología , Infecciones por Actinobacillus/prevención & control , Actinobacillus pleuropneumoniae/clasificación , Actinobacillus pleuropneumoniae/genética , Animales , Anticuerpos Antibacterianos/biosíntesis , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Secuencia Conservada , Evaluación Preclínica de Medicamentos/veterinaria , Ensayo de Inmunoadsorción Enzimática , Femenino , Inmunización Secundaria , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Infiltración Neutrófila/inmunología , Fagocitosis/inmunología , Pleuroneumonía/patología , Pleuroneumonía/prevención & control , Distribución Aleatoria , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Serogrupo , Porcinos , Enfermedades de los Porcinos/inmunología , UDP-Glucosa-Hexosa-1-Fosfato Uridiltransferasa/genética , Vacunación/veterinaria
16.
Pathog Dis ; 76(2)2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29718270

RESUMEN

Group A Streptococcus (GAS) is a globally important human pathogen that causes a broad spectrum of disease ranging from mild superficial infections to severe invasive diseases with high morbidity and mortality. Currently, there is no vaccine available for human use. GAS produces a vast array of virulence factors including multiple adhesin molecules. These mediate binding of the bacteria to host tissues and are essential in the initial phases of infection. Prophylactic vaccination with adhesins is a promising vaccine strategy and many GAS adhesins are currently in development as vaccine candidates. The most advanced candidates, having entered clinical trials, are based on the M protein, while components of the pilus and a number of fibronectin-binding proteins are in pre-clinical development. Adhesin-based vaccines aim to induce protective immunity via two main mechanisms: neutralisation where adhesin-specific antibodies block the ability of the adhesin to bind to host tissue and opsonisation in which adhesin-specific antibodies tag the GAS bacteria for phagocytosis. This review summarises our current knowledge of GAS adhesins and their structural features in the context of vaccine development.


Asunto(s)
Adhesinas Bacterianas/inmunología , Proteínas Bacterianas/inmunología , Infecciones Estreptocócicas/prevención & control , Vacunas Estreptocócicas/inmunología , Vacunas Estreptocócicas/aislamiento & purificación , Streptococcus pyogenes/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Ensayos Clínicos como Asunto , Descubrimiento de Drogas/tendencias , Evaluación Preclínica de Medicamentos , Humanos , Proteínas Opsoninas/sangre
17.
Microb Pathog ; 117: 341-347, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29510207

RESUMEN

The predominant Staphylococcus aureus (S. aureus), an etiological agent of camel mastitis is becoming drug resistant that invites prevention and control strategies. Vaccine production would have a valuable impact on public health. Therefore, in present study, inactivated vaccine with different adjuvants was prepared and evaluated against S. aureus. The vaccinal isolate recovered from camel subclinical mastitis was coagulase positive (PCR based), having expressed pseudocapsule, holding alpha-beta hemolysin characteristics, and multiple drug resistant. Inactivated alum precipitated S. aureus vaccine (APSV) and oil adjuvant S. aureus vaccine (OASV) were prepared after confirming its antigenicity in rabbits. Three groups of rabbits were randomly inoculated with APSV, OASV, and placebo (Unvaccinated, UV). Each group was further divided into two groups based on single and booster dose inoculation. Booster dose of vaccines in rabbits at day 15th of primary inoculation was given. Serum samples were taken on 15, 30, 45 and 60 days of primary inoculation from all rabbits. Analysis of variance was applied to compare geometric mean titer (GMT) of three groups, while t-test was applied to estimate the difference between single and booster dose response. The study found 1010 CFU/mL S. aureus as standard bacterial load for vaccines with higher and sustained antigenicity. The vaccines were safe from morbidity and mortality, and proved effective and stable for 7 and 4 months at 25 °C and 37 °C, respectively. The OASV produced significantly (p < 0.05) higher immune response followed by APSV throughout trial. The highest GMT by APSV and OASV vaccines with single dose inoculation was 37.92 and 69.92 at day 45th post primary inoculation, respectively. Similarly, 59.20 and 142.40 GMTs were noted with booster dose in case of APSV and OASV, respectively. The booster dose presented significantly (p < 0.05) higher GMT than that of single dose inoculation of vaccines. The study concluded APSV and OASV safe, effective, and stable with significant immunogenic results in experimental rabbits.


Asunto(s)
Inmunogenicidad Vacunal/inmunología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/prevención & control , Vacunas Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Vacunación , Vacunas de Productos Inactivados/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Compuestos de Alumbre , Animales , Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/inmunología , Carga Bacteriana , Proteínas Bacterianas/inmunología , Camelus , Coagulasa , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana Múltiple , Femenino , Proteínas Hemolisinas , Inmunización Secundaria , Mastitis/inmunología , Mastitis/microbiología , Mastitis/prevención & control , Aceite Mineral/administración & dosificación , Conejos , Infecciones Estafilocócicas/microbiología , Vacunas Estafilocócicas/administración & dosificación , Staphylococcus aureus/patogenicidad , Factores de Tiempo , Vacunas de Productos Inactivados/administración & dosificación
18.
Hum Vaccin Immunother ; 14(8): 1948-1956, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29543583

RESUMEN

Injection site reactions (ISRs; redness, swelling and pain) commonly occur within 1-2 days after vaccination. After administration of toxoid vaccines including diphtheria toxoid, a later onset of ISRs has also been observed. As the serotype capsular polysaccharides in the 13-valent pneumococcal conjugate vaccine (PCV13) are conjugated to cross-reactive material 197 (CRM197), a nontoxic variant of diphtheria toxin, the onset of ISRs over 14 days was explored in 8 adult studies with 19 cohorts. Subjects received PCV13 with aluminum phosphate (AlPO4, n = 5667) or without AlPO4 (n = 304); 1097 subjects received 23-valent pneumococcal polysaccharide vaccine (PPSV23). Late ISRs with onset between days 6-14 were observed in 8/8 cohorts aged ≥65 years after PCV13 with AlPO4 (incidence across cohorts for redness, 2.3%-19.6%; swelling, 0.9%-10.8%; pain, 1.6%-10.0%) and in 1/1 cohort after PCV13 without AlPO4 (redness 10.5%; swelling 7.5%; pain 12.3%); and in 2/4 cohorts aged 50 to 64 years after PCV13 (redness 3.1%-4.8%; swelling 1.0%-3.2%; pain 3.7%-5%). Late ISRs were not generally observed in 1/1 cohort aged 18 to 49 years after PCV13; in 2/2 cohorts aged ≥53 years after PCV13 revaccination; and in 3/3 cohorts aged ≥60 years who received PPSV23, which does not contain CRM197. Post hoc analysis demonstrated numerically higher pneumococcal immune responses in subgroups with late ISRs versus those without. In conclusion, causality of late ISRs is likely multifactorial, with age and the PCV13 carrier protein CRM197 potentially associated. AlPO4, a vaccine adjuvant, did not appear causally related. Observations do not affect the favorable risk-benefit profile of PCV13.


Asunto(s)
Proteínas Bacterianas/efectos adversos , Reacción en el Punto de Inyección/epidemiología , Infecciones Neumocócicas/prevención & control , Vacunas Neumococicas/efectos adversos , Streptococcus pneumoniae/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/efectos adversos , Adulto , Factores de Edad , Anciano , Compuestos de Aluminio/administración & dosificación , Compuestos de Aluminio/efectos adversos , Proteínas Bacterianas/administración & dosificación , Proteínas Bacterianas/inmunología , Estudios Clínicos como Asunto , Estudios de Cohortes , Humanos , Inmunización Secundaria/efectos adversos , Inmunización Secundaria/métodos , Incidencia , Reacción en el Punto de Inyección/inmunología , Vacunación Masiva/efectos adversos , Vacunación Masiva/métodos , Persona de Mediana Edad , Fosfatos/administración & dosificación , Fosfatos/efectos adversos , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/microbiología , Vacunas Neumococicas/administración & dosificación , Vacunas Neumococicas/inmunología , Medición de Riesgo , Factores de Tiempo , Vacunas Conjugadas/administración & dosificación , Vacunas Conjugadas/efectos adversos , Vacunas Conjugadas/inmunología , Adulto Joven
19.
Vaccine ; 36(8): 1047-1054, 2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29406241

RESUMEN

Pseudomonas aeruginosa is an opportunistic pathogen responsible for a wide range of severe nosocomial and community acquired infections, these infections are major health problems for cystic fibrosis patients and immune-compromised individuals. The emergence of multidrug-resistant isolates highlights the need to develop alternative strategies for treatment of P. aeruginosa infections. Outer membrane vesicles (OMVs) are spherical nanometer-sized proteolipids that are secreted from numerous of pathogenic Gram-negative bacteria, and a number of studies have confirmed the protective efficacy for use of OMVs as candidate vaccines. In this study, OMVs from P. aeruginosa (PA_OMVs) were isolated, formulated with aluminum phosphate adjuvant and used as a vaccine in a mouse model of acute lung infection. The results confirmed that active immunization with PA_OMVs was able to reduce bacterial colonization, cytokine secretion and tissue damage in the lung tissue, thus protecting mice from lethal challenge of P. aeruginosa. Cytokines assay validated that immunization with PA_OMVs was efficient to induce a mixed cellular immune response in mice. Further, high level of specific antibodies was detected in mice immunized with PA_OMVs, and results from opsonophagocytic killing assay and passive immunization suggested that humoral immune response may be critical for PA_OMVs mediated protection. These findings demonstrated that PA_OMVs may be served as a novel candidate vaccine for the prevention of P. aeruginosa infection.


Asunto(s)
Proteínas Bacterianas/inmunología , Inmunidad Humoral/inmunología , Inmunización Pasiva , Proteínas Ligadas a Lípidos/inmunología , Pseudomonas aeruginosa/inmunología , Vacunación , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/uso terapéutico , Compuestos de Aluminio/química , Compuestos de Aluminio/inmunología , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/aislamiento & purificación , Femenino , Inmunidad Celular/inmunología , Proteínas Ligadas a Lípidos/química , Proteínas Ligadas a Lípidos/aislamiento & purificación , Enfermedades Pulmonares/inmunología , Enfermedades Pulmonares/prevención & control , Enfermedades Pulmonares/terapia , Ratones , Ratones Endogámicos BALB C , Fosfatos/química , Fosfatos/inmunología , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/prevención & control , Infecciones por Pseudomonas/terapia
20.
J Microbiol Biotechnol ; 28(1): 157-164, 2018 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-29132197

RESUMEN

Francisella tularensis (FT), a highly infectious pathogen, is considered to be a potential biological weapon owing to the current lack of a human vaccine against it. Tul4 and FopA, both outer membrane proteins of FT, play an important role in the bacterium's immunogenicity. In the present study, we evaluated the immune response of mice-humanized with human CD34+ cells (hu-mice)-to a cocktail of recombinant Tul4 and FopA (rTul4 and rFopA), which were codon-optimized and expressed in Escherichia coli. Not only did the cocktail-immunized hu-mice produce a significant human immunoglobulin response, they also exhibited prolonged survival against an attenuated live vaccine strain as well as human T cells in the spleen. These results suggest that the cocktail of rTul4 and rFopA had successfully induced an immune response in the hu-mice, demonstrating the potential of this mouse model for use in the evaluation of FT vaccine candidates.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Vacunas Bacterianas/inmunología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Francisella tularensis/inmunología , Tularemia/prevención & control , Animales , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Vacunas Bacterianas/administración & dosificación , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Inmunoglobulina G/sangre , Lipoproteínas/genética , Lipoproteínas/inmunología , Ratones , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Análisis de Supervivencia , Linfocitos T/inmunología , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA