Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 297
Filtrar
Más filtros

Medicinas Complementárias
Intervalo de año de publicación
1.
J Biol Chem ; 299(9): 104927, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37330175

RESUMEN

Methicillin-resistant Staphylococcus aureus, or MRSA, is one of the major causative agents of hospital-acquired infections worldwide. Novel antimicrobial strategies efficient against antibiotic-resistant strains are necessary and not only against S. aureus. Among those, strategies that aim at blocking or dismantling proteins involved in the acquisition of essential nutrients, helping the bacteria to colonize the host, are intensively studied. A major route for S. aureus to acquire iron from the host organism is the Isd (iron surface determinant) system. In particular, the hemoglobin receptors IsdH and IsdB located on the surface of the bacterium are necessary to acquire the heme moiety containing iron, making them a plausible antibacterial target. Herein, we obtained an antibody of camelid origin that blocked heme acquisition. We determined that the antibody recognized the heme-binding pocket of both IsdH and IsdB with nanomolar order affinity through its second and third complementary-determining regions. The mechanism explaining the inhibition of acquisition of heme in vitro could be described as a competitive process in which the complementary-determining region 3 from the antibody blocked the acquisition of heme by the bacterial receptor. Moreover, this antibody markedly reduced the growth of three different pathogenic strains of MRSA. Collectively, our results highlight a mechanism for inhibiting nutrient uptake as an antibacterial strategy against MRSA.


Asunto(s)
Anticuerpos Antibacterianos , Staphylococcus aureus Resistente a Meticilina , Receptores de Superficie Celular , Anticuerpos de Dominio Único , Humanos , Antibacterianos/farmacología , Hemo/metabolismo , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Receptores de Superficie Celular/metabolismo , Receptores de Superficie Celular/uso terapéutico , Anticuerpos de Dominio Único/biosíntesis , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/metabolismo , Anticuerpos de Dominio Único/farmacología , Infecciones Estafilocócicas/tratamiento farmacológico , Antígenos Bacterianos/inmunología , Anticuerpos Antibacterianos/genética , Anticuerpos Antibacterianos/inmunología , Camélidos del Nuevo Mundo , Animales , Unión Proteica/efectos de los fármacos , Modelos Moleculares , Simulación de Dinámica Molecular
2.
Sci Rep ; 12(1): 1325, 2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35079054

RESUMEN

Pseudomonas aeruginosa as a common pathogen causing urinary tract infections (UTIs) has been resistant to different antibiotics and developing an effective vaccine can be an alternative strategy. In the present study, the immunogenicity and protection efficacy of formulations composed of a hybrid protein composed of P. aeruginosa V-antigen (PcrV) and exoenzyme S (ExoS) with alum and MPL were evaluated. The hybrid protein could increase the specific systemic and mucosal immune responses, as well as cellular responses as compared with control groups. Combining of alum or MPL adjuvant with the hybrid protein significantly improved the levels of IgG1, serum IgA, mucosal IgG, and IL-17 as compared to the ExoS.PcrV alone. After bladder challenge with a P. aeruginosa strain, the bacterial loads of bladder and kidneys were significantly decreased in mice received ExoS.PcrV admixed with alum and ExoS.PcrV admixed with MPL than controls. The present study indicated that immunization of mice with a hybrid protein composed of ExoS and PcrV could induce multifactorial immune responses and opsonize the bacteria and decrease the viable bacterial cells. Because P. aeruginosa have caused therapeutic challenges worldwide, our study proposed ExoS.PcrV + alum and ExoS.PcrV + MPL as promising candidates for the prevention of infections caused by P. aeruginosa.


Asunto(s)
ADP Ribosa Transferasas/inmunología , Adyuvantes Inmunológicos/farmacología , Antígenos Bacterianos/inmunología , Toxinas Bacterianas/inmunología , Proteínas Citotóxicas Formadoras de Poros/inmunología , Infecciones por Pseudomonas , Pseudomonas aeruginosa/efectos de los fármacos , Animales , Ratones , Ratones Endogámicos BALB C , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/prevención & control
3.
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
4.
Int J Mol Sci ; 22(17)2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34502407

RESUMEN

Mycobacterium tuberculosis, the causative agent of tuberculosis (TB), is a successful intracellular pathogen that is responsible for the highest mortality rate among diseases caused by bacterial infections. During early interaction with the host innate cells, M. tuberculosis cell surface antigens interact with Toll like receptor 4 (TLR4) to activate the nucleotide-binding domain, leucine-rich-repeat containing family, pyrin domain-containing 3 (NLRP3) canonical, and non-canonical inflammasome pathways. NLRP3 inflammasome activation in the alveoli has been reported to contribute to the early inflammatory response that is needed for an effective anti-TB response through production of pro-inflammatory cytokines, including those of the Interleukin 1 (IL1) family. However, overstimulation of the alveolar NLRP3 inflammasomes can induce excessive inflammation that is pathological to the host. Several studies have explored the use of medicinal plants and/or their active derivatives to inhibit excessive stimulation of the inflammasomes and its associated factors, thus reducing immunopathological response in the host. This review describes the molecular mechanism of the NLRP3 inflammasome activation in the alveoli during M. tuberculosis infection. Furthermore, the mechanisms of inflammasome inhibition using medicinal plant and their derivatives will also be explored, thus offering a novel perspective on the alternative control strategies of M. tuberculosis-induced immunopathology.


Asunto(s)
Macrófagos Alveolares/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Tuberculosis/tratamiento farmacológico , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Citocinas/metabolismo , Humanos , Inflamasomas/metabolismo , Inflamación , Interleucina-1beta/metabolismo , Macrófagos Alveolares/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/patogenicidad , Proteína con Dominio Pirina 3 de la Familia NLR/efectos de los fármacos , Plantas Medicinales , Alveolos Pulmonares/patología , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Tuberculosis/metabolismo
5.
Eur J Immunol ; 51(11): 2641-2650, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34398472

RESUMEN

Helicobacter pylori is a Gram-negative bacterium found on the luminal surface of the gastric mucosa in at least 50% of the world's human population. The protective effect of breastfeeding against H. pylori infection has been extensively reported; however, the mechanisms behind this protection remain poorly understood. Human IgA from colostrum has reactivity against H. pylori antigens. Despite that IgA1 and IgA2 display structural and functional differences, their reactivity against H. pylori had not been previously determined. We attested titers and reactivity of human colostrum-IgA subclasses by ELISA, immunoblot, and flow cytometry. Colostrum samples from healthy mothers had higher titers of IgA; and IgA1 mostly recognized H. pylori antigens. Moreover, we found a correlation between IgA1 reactivity and their neutralizing effect determined by inhibition of cytoskeletal changes in AGS cells infected with H. pylori. In conclusion, colostrum-IgA reduces H. pylori infection of epithelial gastric cells, suggesting an important role in preventing the bacteria establishment during the first months of life. As a whole, these results suggest that IgA1 from human colostrum provides protection that may help in the development of the mucosal immune system of newborn children.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Calostro/inmunología , Helicobacter pylori/inmunología , Inmunoglobulina A Secretora/inmunología , Citoesqueleto , Células Epiteliales , Femenino , Mucosa Gástrica/inmunología , Infecciones por Helicobacter/inmunología , Humanos , Embarazo
6.
Int Immunopharmacol ; 96: 107638, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33848909

RESUMEN

The majority of urinary tract infections (UTIs) are caused by uropathogenic Escherichia coli (UPEC). Designing a vaccine will certainly reduce the occurrence of infection and antibiotic resistance of the isolates. Antigen 43 (Ag43) and autotransporter H (UpaH) have been associated with the virulence of UPEC. In the present study, the efficacy of different formulations of a hybrid protein composed of Ag43 and UpaH with and without alum and 1,25(OH)2D3 (Vitamin D3) adjuvants were evaluated in mice model. A significant increase in IgG and cellular responses was developed against Ag43::UpaH as compared to the control mice. The addition of alum or a mixture of alum and Vitamin D3 to the protein significantly enhanced the serum IgG responses and tended to remain in a steady state until 6 months. In addition, the mentioned formulations produced significant amounts of IgG1, IL-4, and IL-17 as compared to the fusion protein alone. In addition to the mentioned formulations, the combination of protein with Vitamin D3 also resulted in significantly higher serum IgA and IFN-γ levels as compared to the fusion protein alone. Mice immunized with fusion plus alum and formulation protein admixed with both alum and Vitamin D3 significantly reduced the bacterial load in the bladders and kidneys of mice as compared to the control. In this study, for the first time, the ability of a novel hybrid protein in combination with adjuvants alum and Vitamin D3 was evaluated against UPEC. Our results indicated that fusion Ag43::UpaH admixed with alum and Vitamin D3 could be a promising candidate against UTIs.


Asunto(s)
Compuestos de Alumbre/farmacología , Colecalciferol/farmacología , Proteínas de Escherichia coli/inmunología , Proteínas Recombinantes de Fusión/inmunología , Infecciones Urinarias/prevención & control , Escherichia coli Uropatógena/inmunología , Factores de Virulencia/inmunología , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/uso terapéutico , Compuestos de Alumbre/administración & dosificación , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/orina , Antígenos Bacterianos/inmunología , Carga Bacteriana/efectos de los fármacos , Carga Bacteriana/inmunología , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/inmunología , Vacunas Bacterianas/aislamiento & purificación , Colecalciferol/administración & dosificación , Citocinas/metabolismo , Inmunidad Humoral/efectos de los fármacos , Inmunización/métodos , Inmunoglobulina G/sangre , Inmunoglobulina G/orina , Inyecciones Intravenosas , Ratones Endogámicos BALB C , Membrana Mucosa/inmunología , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/aislamiento & purificación , Infecciones Urinarias/inmunología
7.
Fish Shellfish Immunol ; 113: 125-138, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33746060

RESUMEN

Oral delivery is the most convenient way to vaccinate cultured fish, however it is still problematic, primarily due to a lack of a commercially valid vaccine vehicle to protect the antigen against gastric degradation and ensure its uptake from the intestine. With the goal of advancing the potential to vaccinate orally, this study evaluates a novel silicon nanoparticle-based vehicle (VacSaf carrier). Aeromonas salmonicida antigens were formulated with the VacSaf carrier using different preparation methods to generate dry powder and liquid formulations. Twelve formulations were first subjected to an in vitro evaluation where the A. salmonicida bacterin conjugated to VacSaf carriers were found superior at inducing pro-inflammatory cytokine expression in primary leucocyte cultures and the macrophage/monocyte cell line RTS-11 compared with A. salmonicida bacterin alone. This was especially apparent after exposure to acid conditions to mimic stomach processing. One formulation (FD1) was taken forward to oral delivery using two doses and two administration schedules (5 days vs 10 days, the latter 5 days on, 5 days off, 5 days on), and the transcript changes of immune genes in the intestine (pyloric caeca, midgut and hindgut) and spleen were evaluated by qPCR and serum IgM was measured by ELISA. The VacSaf carrier alone was shown to be safe for use in vivo, in that no side-effects were seen, but it did induce expression of some cytokines, and may have value as an oral adjuvant candidate. The FD1 bacterin formulation was effective at inducing a range of cytokines associated with innate and adaptive immunity, mainly in the pyloric caeca, compared to A. salmonicida bacterin alone (which had almost no effect), and confirms the immune competence of this gut region following appropriate oral vaccination. These results reveal that in vitro screening of formulations for oral delivery has value and can be used to assess the most promising formulations to test further.


Asunto(s)
Aeromonas salmonicida/inmunología , Vacunas Bacterianas/inmunología , Enfermedades de los Peces/inmunología , Nanopartículas/administración & dosificación , Oncorhynchus mykiss/inmunología , Vacunación/veterinaria , Inmunidad Adaptativa , Adyuvantes Inmunológicos/administración & dosificación , Administración Oral , Animales , Antígenos Bacterianos/inmunología , Vacunas Bacterianas/administración & dosificación , Línea Celular , Sistemas de Liberación de Medicamentos/instrumentación , Sistemas de Liberación de Medicamentos/métodos , Sistemas de Liberación de Medicamentos/veterinaria , Infecciones por Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/veterinaria , Inmunidad Innata , Macrófagos/inmunología , Monocitos/inmunología , Vacunación/instrumentación , Vacunación/métodos
8.
Mol Cell Probes ; 56: 101708, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33636281

RESUMEN

Mesoporous silica nanoparticles (MSNs) have been used in the field of biomedicine as antigen carriers and adjuvants for protective antigens. In the present study, an oral nanovaccine against Vibrio alginolyticus was prepared employing MSNs as carriers. The uptake of the dihydrolipoamide dehydrogenase (DLDH) antigens in the intestine of large yellow croaker was evaluated using an immunohistochemistry assay. Additionally, the effects of the nanovaccine on the early immune response in large yellow croaker were investigated via oral vaccination. The presence of the antigens was detected in the mucosa and lamina propria of the foregut, midgut, and hindgut of large yellow croaker at 3 h following oral immunization. The expression levels of cytokines (i.e., lysozyme, IFN-γ, IFITM, TNF-α, IL-1ß, IL-2, IL-4, IL-10, and IL-13) in the intestine, spleen, and head kidney tissues of large yellow croaker before and after the immune challenge were determined via RT-qPCR assay. The obtained results revealed that the expression levels of lysozyme, IFN-γ, IFITM, TNF-α, IL-1ß, IL-2, IL-4, IL-10, and IL-13 in the intestine and head kidney of the vaccinated large yellow croaker, as well as the expression of lysozyme, IL-1ß, and IL-10 in the spleen, exhibited time-dependent oscillation regulation patterns. Notably, the nanovaccine immunization could induce early (6 h) and high expression of IFN-γ in the spleen and kidney tissues after the bacterial infection. The current study supplements the available data on the early immune response to fish nanovaccines. It also provides a valuable theoretical basis for the future development of large yellow croaker oral vaccines.


Asunto(s)
Antígenos Bacterianos/inmunología , Vacunas Bacterianas/inmunología , Dihidrolipoamida Deshidrogenasa/inmunología , Enfermedades de los Peces/prevención & control , Proteínas de Peces/genética , Vibriosis/veterinaria , Vibrio alginolyticus/inmunología , Administración Oral , Animales , Antígenos Bacterianos/administración & dosificación , Antígenos Bacterianos/genética , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/genética , Dihidrolipoamida Deshidrogenasa/administración & dosificación , Dihidrolipoamida Deshidrogenasa/genética , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/química , Enfermedades de los Peces/genética , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/microbiología , Proteínas de Peces/inmunología , Expresión Génica , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-2/genética , Interleucina-2/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Intestinos/efectos de los fármacos , Intestinos/inmunología , Intestinos/microbiología , Riñón/efectos de los fármacos , Riñón/inmunología , Riñón/microbiología , Muramidasa/genética , Muramidasa/inmunología , Nanopartículas/administración & dosificación , Nanopartículas/química , Perciformes/inmunología , Perciformes/microbiología , Dióxido de Silicio/química , Dióxido de Silicio/inmunología , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/microbiología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Vacunación/métodos , Vibriosis/inmunología , Vibriosis/microbiología , Vibriosis/prevención & control
9.
J Crohns Colitis ; 15(8): 1253-1263, 2021 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-33378524

RESUMEN

BACKGROUND: The role of Mycobacterium avium paratuberculosis [MAP] in inflammatory bowel disease [IBD], especially Crohn's disease [CD] is controversial due conflicting results and lack of reproducibility and standardised tests. The current study focuses on the role of MAP in disease progression and genetic susceptibility, as MAP is likely one of many factors involved in the complex pathogenesis of IBD, potentially affecting a subgroup depending on genetic susceptibility. METHODS: Serum from 812 patients was evaluated with seven immunoglobulin [Ig] isotype-specific serology tests assessing humoral response to three different MAP antigens. For each of these in total 21 tests, the intra-assay and inter-assay coefficients were used to evaluate test accuracy. Reliable assays were subsequently analysed in relation to disease characteristics and need for biologic therapy/surgery. Genome-wide genotyping was available for all participants. Genetic determinants of humoral response to MAP antigens were evaluated using genome-wide association analysis and polygenic risk scores [PRS]. RESULTS: High IgA or IgM response to MAP2609 was associated with increased use of biologic therapy in CD and ulcerative colitis [UC] [odds ratios 2.69; 95% confidence interval 1.44-5.01; and 2.60, 1.46-4.64, respectively]. No associations were seen for risk of surgery [p-values > 0.29]. We could not identify genetic determinants nor polygenic risk scores for MAP response with genome-wide significance. CONCLUSIONS: Extensive assays for serological response to MAP were evaluated using stringent criteria for reliability. Increased IgA and IgM response to MAP antigens was seen in patients exposed to biologic therapy, but no genetic determinants underlying this humoral response were found.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/inmunología , Terapia Biológica , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Mycobacterium avium subsp. paratuberculosis/inmunología , Estudios de Cohortes , Estudios Transversales , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Inmunoglobulina A/sangre , Inmunoglobulina M/sangre , Enfermedades Inflamatorias del Intestino/inmunología , Masculino , Persona de Mediana Edad , Mycobacterium avium subsp. paratuberculosis/genética , Reproducibilidad de los Resultados
10.
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
11.
mSphere ; 5(2)2020 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-32269150

RESUMEN

We are exposed daily to many glycans from bacteria and food plants. Bacterial glycans are generally antigenic and elicit antibody responses. It is unclear if food glycans' sharing of antigens with bacterial glycans influences our immune responses to bacteria. We studied 14 different plant foods for cross-reactivity with monoclonal antibodies (MAbs) against 24 pneumococcal serotypes which commonly cause infections and are included in pneumococcal vaccines. Serotype 15B-specific MAb cross-reacts with fruit peels, and serotype 10A MAb cross-reacts with many natural and processed plant foods. The serotype 10A cross-reactive epitope is terminal 1,6-linked ß-galactose [ßGal(1-6)], present in the rhamno-galacturonan I (RG-I) domain of pectin. Despite wide consumption of pectin, the immune response to 10A is comparable to the responses to other serotypes. An antipectin antibody can opsonize serotype 10A pneumococci, and the shared ßGal(1-6) may be useful as a simple vaccine against 10A. Impact of food glycans should be considered in host-pathogen interactions and future vaccine designs.IMPORTANCE The impact of food consumption on vaccine responses is unknown. Streptococcus pneumoniae (the pneumococcus) is an important human pathogen, and its polysaccharide capsule is used as a vaccine. We show that capsule type 10A in a pneumococcal vaccine shares an antigenic epitope, ßGal(1-6), with pectin, which is in many plant foods and is widely consumed. Immune response to 10A is comparable to that seen with other capsule types, and pectin ingestion may have little impact on vaccine responses. However, antibody to pectin can kill serotype 10A pneumococci and this shared epitope may be considered in pneumococcal vaccine designs.


Asunto(s)
Antígenos Bacterianos/inmunología , Cápsulas Bacterianas/inmunología , Reacciones Cruzadas , Pectinas/inmunología , Streptococcus pneumoniae/inmunología , Anticuerpos Monoclonales/inmunología , Epítopos/inmunología , Frutas , Humanos , Fagocitosis , Serogrupo , Verduras
12.
Res Vet Sci ; 130: 1-10, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32105948

RESUMEN

On a spring calving, pastoral dairy farm, the first 40 heifer calves born after calving mid-point (50% of the herd calved) were blood sampled within 24 h. Thirty were selected, using stratified randomisation to form two equal groups (treatment and control) with the same distribution of serum total protein, copper, selenium, zinc, and manganese concentrations, age and breed. From the remaining 10 calves, five were randomly selected into a sentinel group to assess field exposure to Salmonella spp. All calves received two injections of a killed vaccine containing Salmonella spp. antigens at 2 and 6 weeks of age. Concurrently, the treatment group were injected with 1 mL/50 kg trace mineral supplement (TMS) containing 40 mg zinc, 10 mg manganese, 5 mg selenium, 15 mg copper per mL. Sentinel animals received no injections. All animals were bled from 2 to 9 weeks for assay of immune function. At three and four weeks, white blood cells from TMS calves had an increased percentage of cells phagocytosing (effect size = 9.36 and 4.35) and increased number of bacteria ingested per cell (effect size = 0.93 and 1.52). No differences were detected in gamma interferon response (effect size <0.15) or Salmonella sp. antibody titres (effect size <0.20).


Asunto(s)
Antígenos Bacterianos/inmunología , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Salmonella/fisiología , Oligoelementos/metabolismo , Alimentación Animal/análisis , Animales , Animales Recién Nacidos , Bovinos , Dieta/veterinaria , Suplementos Dietéticos/análisis , Femenino , Inyecciones Subcutáneas/veterinaria , Distribución Aleatoria , Oligoelementos/administración & dosificación , Vacunas de Productos Inactivados/administración & dosificación
13.
Mol Pharm ; 16(10): 4372-4386, 2019 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-31437396

RESUMEN

Cationic liposomes prepared from dimethyldioctadecylammonium bromide (DDAB) and trehalose 6,6'-dibehenate (TDB) are strong liposomal adjuvants. As with many liposome formulations, within the laboratory DDAB:TDB is commonly prepared by the thin-film method, which is difficult to scale-up and gives high batch-to-batch variability. In contrast, controllable technologies such as microfluidics offer robust, continuous, and scale-independent production. Therefore, within this study, we have developed a microfluidic production method for cationic liposomal adjuvants that is scale-independent and produces liposomal adjuvants with analogous biodistribution and immunogenicity compared to those produced by the small-scale lipid hydration method. Subsequently, we further developed the DDAB:TDB adjuvant system to include a lymphatic targeting strategy using microfluidics. By exploiting a biotin-avidin complexation strategy, we were able to manipulate the pharmacokinetic profile and enhance targeting and retention of DDAB:TDB and antigen within the lymph nodes. Interestingly, redirecting these cationic liposomal adjuvants did not translate into notably improved vaccine efficacy.


Asunto(s)
Adyuvantes Inmunológicos/química , Cationes/química , Liposomas/química , Ganglios Linfáticos/efectos de los fármacos , Microfluídica , Compuestos de Amonio Cuaternario/química , Vacunas contra la Tuberculosis/administración & dosificación , Adyuvantes Inmunológicos/administración & dosificación , Animales , Antígenos Bacterianos/administración & dosificación , Antígenos Bacterianos/inmunología , Femenino , Inmunización , Liposomas/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Distribución Tisular , Tuberculosis/inmunología , Tuberculosis/prevención & control , Vacunas contra la Tuberculosis/inmunología , Vacunas contra la Tuberculosis/farmacocinética
14.
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
15.
Sci Rep ; 9(1): 8931, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31222043

RESUMEN

Campylobacter infections sourced mainly to poultry products, are the most important bacterial foodborne zoonoses worldwide. No effective measures to control these infections in broiler production exist to date. Here, we used passive immunization with hyperimmune egg yolks to confer broad protection of broilers against Campylobacter infection. Two novel vaccines, a bacterin of thirteen Campylobacter jejuni (C. jejuni) and C. coli strains and a subunit vaccine of six immunodominant Campylobacter antigens, were used for the immunization of layers, resulting in high and prolonged levels of specific immunoglobulin Y (IgY) in the hens' yolks. In the first in vivo trial, yolks (sham, bacterin or subunit vaccine derived) were administered prophylactically in the broiler feed. Both the bacterin- and subunit vaccine-induced IgY significantly reduced the number of Campylobacter-colonized broilers. In the second in vivo trial, the yolks were administered therapeutically during three days before euthanasia. The bacterin IgY resulted in a significant decrease in C. jejuni counts per infected bird. The hyperimmune yolks showed strong reactivity to a broad representation of C. jejuni and C. coli clonal complexes. These results indicate that passive immunization with hyperimmune yolks, especially bacterin derived, offers possibilities to control Campylobacter colonization in poultry.


Asunto(s)
Alimentación Animal , Anticuerpos Antibacterianos/inmunología , Campylobacter jejuni/aislamiento & purificación , Pollos/microbiología , Suplementos Dietéticos , Yema de Huevo/inmunología , Animales , Antígenos Bacterianos/inmunología , Campylobacter jejuni/crecimiento & desarrollo , Campylobacter jejuni/inmunología , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Heces/microbiología , Inmunización Pasiva
16.
mSphere ; 4(3)2019 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-31217301

RESUMEN

Inhalational anthrax caused by Bacillus anthracis, a spore-forming Gram-positive bacterium, is a highly lethal infection. Antibodies targeting the protective antigen (PA) binding component of the toxins have recently been authorized as an adjunct to antibiotics, although no conclusive evidence demonstrates that anthrax antitoxin therapy has any significant benefit. We discuss here the rational basis of anti-PA development regarding the pathogenesis of the disease. We argue that inductive reasoning may induce therapeutic bias. We identified anthrax animal model analysis as another bias. Further studies are needed to assess the benefit of anti-PA antibodies in the treatment of inhalational anthrax, while a clearer consensus should be established around what evidence should be proven in an anthrax model.


Asunto(s)
Carbunco/inmunología , Carbunco/terapia , Anticuerpos Antibacterianos/uso terapéutico , Bacillus anthracis/inmunología , Inmunoterapia , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/terapia , Animales , Anticuerpos Monoclonales/uso terapéutico , Antígenos Bacterianos/inmunología , Antitoxinas/uso terapéutico , Toxinas Bacterianas/inmunología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos
17.
PLoS One ; 14(3): e0212946, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30845147

RESUMEN

Campylobacter jejuni is a zoonotic agent responsible for the foodborne gastroenteritis campylobacteriosis. Control of C. jejuni load in the poultry primary production is recognized as an avenue to reduce human exposure to the pathogen. As for now, no commercially applicable control methods exist at the farm. Several studies tested egg yolk powders, potentiated or not against C. jejuni, as feed additives for chicken and suggested that the quantity and quality of the antibodies presence in the yolk are determinant factors for the full success of this approach. Unfortunately, data from these studies inconsistently showed a reduction of cecal C. jejuni carriage. Our first goal wwas to characterize (quantification by ELISA, agglutination test, bacterial antigen recognition profiles by Western blot, bactericidal effect by serum killing assays and C. jejuni mobility by soft agar migation) the antibodies extracted from egg yolk powders originating from different egg production protocols. Secondly, these powders were microencapsulated and recharacterized. Finally the protected powders were tested as a feed additive to destabilize C. jejuni colonization in an in vivo assay. Despite the in vitro results indicating the ability of the egg yolk powders to recognize Campylobacter and potentially alter its colonization of the chicken caecum, these results were not confirmed in the in vivo trial despite that specific caecal IgY directed toward Campylobacter were detected in the groups receiving the protected powders. More research is needed on Campylobacter in order to effectively control this pathogen at the farm.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Infecciones por Campylobacter/prevención & control , Campylobacter jejuni/inmunología , Yema de Huevo/inmunología , Aditivos Alimentarios/administración & dosificación , Alimentación Animal , Animales , Anticuerpos Antibacterianos/administración & dosificación , Antígenos Bacterianos/inmunología , Carga Bacteriana/efectos de los fármacos , Infecciones por Campylobacter/microbiología , Infecciones por Campylobacter/veterinaria , Campylobacter jejuni/aislamiento & purificación , Ciego/microbiología , Pollos/microbiología , Composición de Medicamentos , Evaluación Preclínica de Medicamentos , Enfermedades Transmitidas por los Alimentos/microbiología , Enfermedades Transmitidas por los Alimentos/prevención & control , Inmunoglobulinas/administración & dosificación , Inmunoglobulinas/inmunología , Productos Avícolas/envenenamiento , Polvos , Resultado del Tratamiento
18.
Mar Drugs ; 17(3)2019 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-30889882

RESUMEN

In this study, the low-molecular-weight (LMW) fucoidan, rich in fucose and sulfate, was extracted and purified from the edible brown seaweed, Laminaria japonica. In this study, we orally administered LMW fucoidan to mice for 6 weeks. We then examined fucoidan's effects on innate immunity, adaptive immunity, and Mycoplasma pneumoniae (MP)-antigen-stimulated immune responses. Our data showed that LMW fucoidan stimulated the innate immune system by increasing splenocyte proliferation, natural killer (NK) cell activity, and phagocytic activity. LMW fucoidan also increased interleukin (IL)-2, IL-4, and interferon (IFN)-γ secretion by splenocytes and immunoglobulin (Ig)-G and IgA content in serum, which help regulate adaptive immune cell functions, and decreased allergen-specific IgE. In MP-antigen-stimulated immune responses, the IgM and IgG content in the serum were significantly higher in the LMW fucoidan group after MP-antigen stimulation. Our study provides further information about the immunomodulatory effects of LMW fucoidan and highlights a potential role in preventing M. pneumoniae infection.


Asunto(s)
Suplementos Dietéticos , Neumonía por Mycoplasma/prevención & control , Polisacáridos/administración & dosificación , Sustancias Protectoras/administración & dosificación , Inmunidad Adaptativa/efectos de los fármacos , Animales , Antígenos Bacterianos/administración & dosificación , Antígenos Bacterianos/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunidad Innata/efectos de los fármacos , Laminaria/química , Ratones , Ratones Endogámicos BALB C , Peso Molecular , Mycoplasma pneumoniae/inmunología , Neumonía por Mycoplasma/inmunología , Neumonía por Mycoplasma/microbiología , Polisacáridos/química , Sustancias Protectoras/química
19.
Invest Ophthalmol Vis Sci ; 60(1): 420-429, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30695094

RESUMEN

Purpose: We determine the changes in intestinal microbiota and/or disruptions in intestinal homeostasis during uveitis. Methods: Experimental autoimmune uveitis (EAU) was induced in B10.RIII mice with coadministration of interphotoreceptor retinoid-binding protein peptide (IRBP) and killed mycobacterial antigen (MTB) as an adjuvant. Using 16S rRNA gene sequencing, we looked at intestinal microbial differences during the course of uveitis, as well as intestinal morphologic changes, changes in intestinal permeability by FITC-dextran leakage, antimicrobial peptide expression in the gastrointstinal tract, and T lymphocyte prevalence before and at peak intraocular inflammation. Results: We demonstrate that increased intestinal permeability and antimicrobial peptide expression in the intestinal tract coincide in timing with increased effector T cells in the mesenteric lymph nodes, during the early stages of uveitis, before peak inflammation. Morphologic changes in the intestine were most prominent during this phase, but also occurred with adjuvant MTB alone, whereas increased intestinal permeability was found only in IRBP-immunized mice that develop uveitis. We also demonstrate that the intestinal microbiota were altered during the course of uveitis, and that some of these changes are specific to uveitic animals, whereas others are influenced by adjuvant MTB alone. Intestinal permeability peaked at 2 weeks, coincident with an increase in intestinal bacterial strain differences, peak lipocalin production, and peak uveitis. Conclusions: An intestinal dysbiosis accompanies a disruption in intestinal homeostasis in autoimmune uveitis, although adjuvant MTB alone promotes intestinal disruption as well. This may indicate a novel axis for future therapeutic targeting experimentally or clinically.


Asunto(s)
Enfermedades Autoinmunes/microbiología , Disbiosis/microbiología , Microbioma Gastrointestinal/inmunología , Homeostasis/fisiología , Intestinos/fisiología , Uveítis/microbiología , Animales , Antígenos Bacterianos/inmunología , Enfermedades Autoinmunes/inmunología , Ensayo de Inmunoadsorción Enzimática , Proteínas del Ojo , Citometría de Flujo , Lipocalinas/metabolismo , Ratones , Ratones Mutantes , Modelos Animales , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/inmunología , ARN Ribosómico 16S/genética , Proteínas de Unión al Retinol , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Subgrupos de Linfocitos T/inmunología , Uveítis/inmunología , Proteína de la Zonula Occludens-1/metabolismo
20.
Sci Rep ; 9(1): 715, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30679470

RESUMEN

Sublingual immunization is emerging as an alternative to nasal immunization and induction of mucosal IgA responses. Using Bacillus anthracis edema toxin (EdTx) as an adjuvant, we previously showed that innate responses triggered after sublingual immunization could limit generation of IgA responses. We tested whether co-administration of a neutrophil elastase inhibitor (NEI) could rescue the ability of EdTx to induce broad antibody responses, including mucosal IgA. NEI supplementation of sublingual vaccines containing EdTx promoted antigen-specific serum IgA responses but also enhanced serum IgG1, and IgG2b responses. This enhancing effect of NEI did not extend to all antibody isotypes and IgG sublclasses, since NEI  reduced serum IgE responses and did not affect IgG2a/c and IgG3 responses. NEI supplementation also promoted anti-Bacillus anthracis protective antigen (PA) neutralizing antibodies and enhanced high affinity IgG1 and IgA antibodies. In addition to serum IgA, NEI supplementation stimulated antigen-specific mucosal IgA responses in the GI tract, and enhanced antigen-specific IgG responses in vaginal washes. Analysis of CD4+ T helper cell responses revealed that co-administration of NEI broadened the profile of cytokine responses, by stimulating Th1, Th2, Th17, and Tfh cytokines. We also noted that NEI had a higher stimulatory effect on IL-5, IL-10, IL-17 responses.


Asunto(s)
Formación de Anticuerpos/inmunología , Antígenos Bacterianos/inmunología , Toxinas Bacterianas/inmunología , Suplementos Dietéticos , Inmunidad Mucosa/inmunología , Membrana Mucosa/inmunología , Proteínas Inhibidoras de Proteinasas Secretoras/administración & dosificación , Vacunas/administración & dosificación , Adyuvantes Inmunológicos , Administración Sublingual , Animales , Antígenos Bacterianos/administración & dosificación , Toxinas Bacterianas/administración & dosificación , Femenino , Inmunoglobulina A/sangre , Inmunoglobulina A/inmunología , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Ratones , Ratones Endogámicos C57BL , Linfocitos T Colaboradores-Inductores , Vacunación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA