Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(16)2024 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-39201440

RESUMEN

Th1 and Th2 cytokines determine the outcome of Leishmania major infection and immune protection depends mainly on memory T cells induced during vaccination. This largely hinges on the nature and type of memory T cells produced. In this study, transgenic Leishmania major strains expressing membrane-associated ovalbumin (mOVA) and soluble ovalbumin (sOVA) were used as a model to study whether fully differentiated Th1/Th2 and Th17 cells can recall immune memory and tolerate pathogen manipulation. Naïve OT-II T cells were polarised in vitro into Th1/Th2 cells, and these cells were transferred adoptively into recipient mice. Following the transferral of the memory cells, the recipient mice were challenged with OVA transgenic Leishmania major and a wild-type parasite was used a control. The in vitro-polarised T helper cells continued to produce the same cytokine signatures after being challenged by both forms of OVA-expressing Leishmania major parasites in vivo. This suggests that antigen-experienced cells remain the same or unaltered in the face of OVA-transgenic Leishmania major. Such ability of these antigen-experienced cells to remain resilient to manipulation by the parasite signifies that vaccines might be able to produce immune memory responses and defend against parasitic immune manipulation in order to protect the host from infection.


Asunto(s)
Memoria Inmunológica , Leishmania major , Ovalbúmina , Células TH1 , Células Th17 , Células Th2 , Animales , Leishmania major/inmunología , Ovalbúmina/inmunología , Ratones , Células TH1/inmunología , Células Th2/inmunología , Células Th17/inmunología , Citocinas/metabolismo , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/parasitología , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Femenino , Ratones Transgénicos
2.
Int J Mol Sci ; 23(3)2022 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-35163381

RESUMEN

Schistosoma mansoni uses different mechanisms to escape its host's immunity. Understanding the ability of memory T cells to withstand this pathogen's manipulation is important for the development of effective vaccines against this immunomodulatory pathogen. In this study, ovalbumin (OVA) transgenic S. mansoni is used as a tool to investigate whether fully differentiated Th1, Th2 and Th17 cells are able to withstand pathogen manipulation. Naïve T cells from OT-II T cell receptor transgenic mice with a specificity for OVA were differentiated into Th1, Th2, and Th17 polarised memory cells in vitro. These cells were adoptively transferred into recipient mice to investigate whether these polarised immune memory T cells are resilient in the face of pathogen-mediated manipulation. After transferring memory cells, mice were challenged with OVA-transduced S. mansoni eggs as well as wild-type controls. The in vitro differentiated Th1, Th2 and Th17 memory cells continued to produce the same cytokines when challenged by OVA-expressing S. mansoni eggs as to these they produced when transferred in vivo, suggesting that the Th phenotypes of the memory T cells remains unaltered in the face of stimulation by S. mansoni. The ability of memory T cells to remain resilient to manipulation by the parasite suggests that vaccines might be able to produce immune memory responses able to withstand S. mansoni immune manipulation and hence protect the host from infection.


Asunto(s)
Inmunidad , Schistosoma mansoni/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Traslado Adoptivo , Animales , Antígenos/inmunología , Polaridad Celular , Proliferación Celular , Citocinas/metabolismo , Femenino , Memoria Inmunológica , Ganglios Linfáticos/metabolismo , Ratones Endogámicos BALB C , Ratones Transgénicos , Ovalbúmina/inmunología , Óvulo/metabolismo , Esquistosomiasis mansoni/inmunología , Bazo/metabolismo , Células TH1/inmunología , Células Th17/inmunología , Células Th2/inmunología
3.
Artículo en Inglés | MEDLINE | ID: mdl-32793510

RESUMEN

The cell surface mucin MUC1 is an important host factor limiting Helicobacter pylori (H. pylori) pathogenesis in both humans and mice by providing a protective barrier and modulating mucosal epithelial and leukocyte responses. The aim of this study was to establish the time-course of molecular events in MUC1-modulated gene expression profiles in response to H. pylori infection in wild type (WT) and MUC1-deficient mice using microarray-determined mRNA expression, gene network analysis and Ingenuity Pathway Analysis (IPA). A time-course over the first 72 h of infection showed significantly higher mucosal loads of bacteria at 8 h of infection in Muc1-/- mice compared with WT, confirming its importance in the early stages of infection (P = 0.0003). Microarray analysis revealed 266 differentially expressed genes at one or more time-points over 72 h in the gastric mucosa of Muc1-/- mice compared with WT control using a threshold of 2-fold change. The SPINK1 pancreatic cancer canonical pathway was strongly inhibited in Muc1-/- mice compared with WT at sham and 8 h infection (P = 6.08E-14 and P = 2.25 E-19, respectively) but potently activated at 24 and 72 h post-infection (P = 1.38E-22 and P = 5.87E-13, respectively). The changes in this pathway are reflective of higher expression of genes mediating digestion and absorption of lipids, carbohydrates, and proteins at sham and 8 h infection in the absence of MUC1, but that this transcriptional signature is highly down regulated as infection progresses in the absence of MUC1. Uninfected Muc1-/- gastric tissue was highly enriched for expression of factors involved in lipid metabolism and 8 h infection further activated this network compared with WT. As infection progressed, a network of antimicrobial and anti-inflammatory response genes was more highly activated in Muc1-/- than WT mice. Key target genes identified by time-course microarrays were independently validated using RT-qPCR. These results highlight the dynamic interplay between the host and H. pylori, and the role of MUC1 in host defense, and provide a general picture of changes in cellular gene expression modulated by MUC1 in a time-dependent manner in response to H. pylori infection.


Asunto(s)
Mucosa Gástrica , Infecciones por Helicobacter , Mucina-1/genética , Animales , Helicobacter pylori , Ratones , Transcriptoma
4.
Exp Parasitol ; 206: 107767, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31520603

RESUMEN

Schistosoma mansoni eggs can influence immune responses directed at them, and the mechanisms by which this is achieved are being unravelled. Going towards, developing effective tools for the study of how S. mansoni influences naïve T cells, we have developed S. mansoni eggs expressing chicken ovalbumin (OVA), using a lentiviral transduction system. Indeed, such a parasite could be used in conjunction with cells from OT-II transgenic mice as a source of naïve, antigen-specific T cells. The expression of the transgenic protein was confirmed by real-time RT-PCR of OVA-specific mRNA and western blotting using polyclonal antibodies specific for OVA. T cells from OT-II transgenic mice expressing a T cell receptor specific for the OVA323-339 peptide recognised the OVA-transduced S. mansoni eggs. Using flow cytometry on CFSE-labelled OT-II splenocytes, we demonstrated that OVA-transduced eggs elicit higher OT-II proliferative responses than untransduced eggs. The OT-II T cells also produced TNF-α and IFN-γ following exposure to OVA-transduced eggs. In addition, moderate amounts of IL-6 and IL-17A were also detected. In contrast, no IL-10, IL-4 and IL-2 were detected in cultures, whether the cells were stimulated with transduced or untransduced eggs. Thus, the cytokine signatures showed the transfected eggs induced predominantly a Th1 response, with a small amount of IL-6 and IL-17.


Asunto(s)
Ovalbúmina/análisis , Receptores de Antígenos de Linfocitos T/inmunología , Schistosoma mansoni/metabolismo , Linfocitos T/inmunología , Animales , Western Blotting , Pollos , Citocinas/análisis , Citocinas/metabolismo , Electroforesis en Gel de Agar , Femenino , Citometría de Flujo , Interleucina-17/análisis , Interleucina-17/metabolismo , Interleucina-2/análisis , Interleucina-2/metabolismo , Interleucina-6/análisis , Interleucina-6/metabolismo , Hígado/parasitología , Ganglios Linfáticos/citología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Ovalbúmina/genética , Ovalbúmina/inmunología , Ovalbúmina/metabolismo , Óvulo/metabolismo , ARN Mensajero/análisis , ARN Mensajero/aislamiento & purificación , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Antígenos de Linfocitos T/genética , Transcripción Reversa , Schistosoma mansoni/genética , Schistosoma mansoni/crecimiento & desarrollo , Bazo/citología , Linfocitos T/citología
5.
Parasit Vectors ; 12(1): 303, 2019 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-31202271

RESUMEN

Parasites comprise diverse and complex organisms, which substantially impact human and animal health. Most parasites have complex life-cycles, and by virtue of co-evolution have developed multifaceted, often life-cycle stage-specific relationships with the immune system of their hosts. The complexity in the biology of many parasites often limits our knowledge of parasite-specific immune responses, to in vitro studies only. The relatively recent development of methods to stably manipulate the genetic make-up of many parasites has allowed a better understanding of host-parasite interactions, particularly in vivo. In this regard, the use of transgenic parasites can facilitate the study of immunomodulatory mechanisms under in vivo conditions. Therefore, in this review, we specifically highlighted the current developments in the use of transgenic parasites to unravel the host's immune response to different life-cycle stages of some key parasite species such as Leishmania, Schistosoma, Toxoplasma, Plasmodium and Trypanosome and to some degree, the use of transgenic nematode parasites is also briefly discussed.


Asunto(s)
Técnicas de Transferencia de Gen , Interacciones Huésped-Parásitos/inmunología , Parásitos/genética , Parásitos/inmunología , Animales , Interacciones Huésped-Parásitos/genética , Humanos , Leishmania/genética , Leishmania/inmunología , Estadios del Ciclo de Vida/genética , Estadios del Ciclo de Vida/inmunología , Ratones , Plasmodium/genética , Plasmodium/inmunología , Toxoplasma/genética , Toxoplasma/inmunología
6.
J Immunol ; 200(5): 1901-1916, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29378910

RESUMEN

Mucosal-associated invariant T (MAIT) cells produce inflammatory cytokines and cytotoxic granzymes in response to by-products of microbial riboflavin synthesis. Although MAIT cells are protective against some pathogens, we reasoned that they might contribute to pathology in chronic bacterial infection. We observed MAIT cells in proximity to Helicobacter pylori bacteria in human gastric tissue, and so, using MR1-tetramers, we examined whether MAIT cells contribute to chronic gastritis in a mouse H. pylori SS1 infection model. Following infection, MAIT cells accumulated to high numbers in the gastric mucosa of wild-type C57BL/6 mice, and this was even more pronounced in MAIT TCR transgenic mice or in C57BL/6 mice where MAIT cells were preprimed by Ag exposure or prior infection. Gastric MAIT cells possessed an effector memory Tc1/Tc17 phenotype, and were associated with accelerated gastritis characterized by augmented recruitment of neutrophils, macrophages, dendritic cells, eosinophils, and non-MAIT T cells and by marked gastric atrophy. Similarly treated MR1-/- mice, which lack MAIT cells, showed significantly less gastric pathology. Thus, we demonstrate the pathogenic potential of MAIT cells in Helicobacter-associated immunopathology, with implications for other chronic bacterial infections.


Asunto(s)
Gastritis/inmunología , Infecciones por Helicobacter/inmunología , Helicobacter pylori/inmunología , Células T Invariantes Asociadas a Mucosa/inmunología , Adulto , Animales , Línea Celular Tumoral , Femenino , Mucosa Gástrica/inmunología , Humanos , Memoria Inmunológica/inmunología , Células Jurkat , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Linfocitos T Citotóxicos/inmunología
7.
Helicobacter ; 23(1)2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29235197

RESUMEN

BACKGROUND: Helicobacter pylori has undergone considerable adaptation to allow chronic persistence within the gastric environment. While H. pylori-associated diseases are driven by an excessive inflammation, severe gastritis is detrimental to colonization by this pathogen. Hence, H. pylori has developed strategies to minimize the severity of gastritis it triggers in its host. Superoxide dismutase (SOD) is well known for its role in protecting against oxidative attack; less recognized is its ability to inhibit immunity, shown for SOD from mammalian sources and those of some bacterial species. This study examined whether H. pylori SOD (HpSOD) has the ability to inhibit the host immune response to these bacteria. MATERIALS AND METHODS: The ability of recombinant HpSOD to modify the response to LPS was measured using mouse macrophages. A monoclonal antibody against HpSOD was generated and injected into H. pylori-infected mice. RESULTS: Addition of HpSOD to cultures of mouse macrophages significantly inhibited the pro-inflammatory cytokine response to LPS stimulation. A monoclonal antibody was generated that was specific for SOD from H. pylori. When injected into mice infected with H. pylori for 3 months, this antibody was readily detected in both sera and gastric tissues 5 days later. While treatment with anti-HpSOD had no effect on H. pylori colonization at this time point, it significantly increased the levels of a range of pro-inflammatory cytokines in the gastric tissues. This did not occur with antibodies against other antioxidant enzymes. CONCLUSIONS: SOD from H. pylori can inhibit the production of pro-inflammatory cytokine during in vivo infection.


Asunto(s)
Citocinas/inmunología , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/enzimología , Interacciones Huésped-Patógeno/inmunología , Superóxido Dismutasa/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/metabolismo , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Femenino , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/metabolismo , Mediadores de Inflamación/inmunología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Especificidad de la Especie , Superóxido Dismutasa/antagonistas & inhibidores
8.
Int J Biochem Cell Biol ; 92: 95-103, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28951199

RESUMEN

Activation of protease-activated receptor-2 (PAR2) expressed by T cells has been linked to the bone loss associated with periodontitis. We generated PAR2 conditional-null mice and crossed these with mice expressing Cre recombinase under control of the Lck proximal promoter, to produce T cell-specific PAR2-null mice in order to further study the cellular mechanism involved in periodontitis. Here we report that efficient deletion of PAR2 in thymocytes isolated from T cell-specific PAR2-null mice resulted in thymic and splenic hypoplasia and a reduction in the cells of the cortex and a loss of distinction between the cortex and the medulla of the thymus. FACS analysis confirmed significant reductions in CD4 and CD8 double negative (DN3 and DN4) sub-populations, as well as double positive and single positive T cells, in T cell-specific PAR2-null mice compared to Cre expressing PAR2 wild-type mice. The proportion of annexin V positive and propidium iodide negative cells was increased in CD4 and CD8 double negative, double positive and single positive T cells from T cell-specific PAR2-null mice. No change in the proportion of Ki67 positive cells was observed in sections of thymus from T cell-specific PAR2-null mice, suggesting that the depletion of T cell sub-populations in T cell-specific PAR2-null mice resulted from increased apoptosis rather than reduced proliferation. Together, these results demonstrate that PAR2 plays an important and previously unrecognised anti-apoptotic role in T cell development and suggest that the PAR2 conditional-null mouse will be an important resource for determining tissue and cell specific effects of PAR2.


Asunto(s)
Receptor PAR-2/deficiencia , Receptor PAR-2/genética , Linfocitos T/citología , Linfocitos T/metabolismo , Animales , Apoptosis/genética , Hiperplasia/genética , Activación de Linfocitos , Ratones , Ratones Noqueados , Bazo/inmunología , Bazo/patología , Linfocitos T/inmunología , Timo/inmunología , Timo/patología
9.
Gut ; 65(7): 1087-99, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26079943

RESUMEN

OBJECTIVES: The mucin MUC1, best known for providing an epithelial barrier, is an important protective host factor in both humans and mice during Helicobacter pylori pathogenesis. This study aimed to identify the long-term consequences of MUC1 deficiency on H. pylori pathogenesis and the mechanism by which MUC1 protects against H. pylori gastritis. DESIGN: Wildtype and Muc1(-/-) mice were infected for up to 9 months, and the gastric pathology, immunological response and epigenetic changes assessed. The effects of MUC1 on the inflammasome, a potent inflammatory pathway, were examined in macrophages and H. pylori-infected mice deficient in both MUC1 and inflammasome components. RESULTS: Muc1(-/-) mice began to die 6 months after challenge, indicating Muc1 deficiency made H. pylori a lethal infection. Surprisingly, chimaeric mouse infections revealed MUC1 expression by haematopoietic-derived immune cells limits H. pylori-induced gastritis. Gastritis in infected Muc1(-/-) mice was associated with elevated interleukin (IL)-1ß and epigenetic changes in their gastric mucosa similar to those in transgenic mice overexpressing gastric IL-1ß, implicating MUC1 regulation of an inflammasome. In support of this, infected Muc1(-/-)Casp1(-/-) mice did not develop severe gastritis. Further, MUC1 regulated Nlrp3 expression via an nuclear factor (NF)-κB-dependent pathway and reduced NF-κB pathway activation via inhibition of IRAK4 phosphorylation. The importance of this regulation was proven using Muc1(-/-)Nlrp3(-/-) mice, which did not develop severe gastritis. CONCLUSIONS: MUC1 is an important, previously unidentified negative regulator of the NLRP3 inflammasome. H. pylori activation of the NLRP3 inflammasome is normally tightly regulated by MUC1, and loss of this critical regulation results in the development of severe pathology.


Asunto(s)
Gastritis/microbiología , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/patogenicidad , Inflamasomas/metabolismo , Mucina-1/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Caspasa 1/genética , Metilación de ADN , Femenino , Mucosa Gástrica/inmunología , Mucosa Gástrica/metabolismo , Gastritis/patología , Expresión Génica , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/inmunología , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Mucina-1/genética , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Transducción de Señal , Factores de Tiempo , Factor Trefoil-2/genética
10.
Expert Rev Vaccines ; 14(4): 579-88, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25591728

RESUMEN

The early stages of the induction of a primary immune response to a vaccine can shape the overall quality of the immune memory generated and hence affect the success of the vaccine. This early interaction between a vaccine and the immune system occurs first at the site of vaccination and can be explored using afferent cannulation. Subsequently, the vaccine and adjuvant activates the local draining lymph node. These interactions can be studied in real time in vivo using efferent lymphatic duct cannulation in large animal models and are the subject of this review. Depending on how the vaccine is delivered, the draining lymph nodes of different organs can be accessed, facilitating the testing of tissue-specific vaccinations. The efferent lymphatic cannulation model provides an avenue to study the effect of both adjuvants and antigen on the local immune system, and hence opens a pathway toward developing more effective ways of inducing immunity.


Asunto(s)
Cateterismo , Inmunidad Activa , Vasos Linfáticos/fisiología , Vacunas/administración & dosificación , Vacunas/inmunología , Animales , Modelos Animales
11.
Nat Commun ; 5: 5375, 2014 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-25400038

RESUMEN

Schistosomiasis, one of the most important neglected tropical diseases worldwide, is caused by flatworms (blood flukes or schistosomes) that live in the bloodstream of humans. The hepatointestinal form of this debilitating disease results from a chronic infection with Schistosoma mansoni or Schistosoma japonicum. No vaccine is available to prevent schistosomiasis, and treatment relies predominantly on the use of a single drug, praziquantel. In spite of considerable research effort over the years, very little is known about the complex in vivo events that lead to granuloma formation and other pathological changes during infection. Here we use, for the first time, a lentivirus-based transduction system to deliver microRNA-adapted short hairpin RNAs (shRNAmirs) into the parasite to silence and explore selected protein-encoding genes of S. mansoni implicated in the disease process. This gene-silencing system has potential to be used for functional genomic-phenomic studies of a range of socioeconomically important pathogens.


Asunto(s)
Antígenos Helmínticos/genética , Proteínas del Huevo/genética , Técnicas de Silenciamiento del Gen/métodos , Granuloma/parasitología , Lentivirus/genética , Schistosoma mansoni/virología , Esquistosomiasis mansoni/virología , Transducción Genética/métodos , Animales , Antígenos Helmínticos/biosíntesis , Proteínas del Huevo/biosíntesis , Huevos/virología , Silenciador del Gen , Granuloma/prevención & control , Ratones Endogámicos BALB C , MicroARNs , ARN Interferente Pequeño/genética , Esquistosomiasis mansoni/patología
12.
PLoS One ; 9(5): e96563, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24794215

RESUMEN

Alphaherpesviruses, which have co-evolved with their hosts for more than 200 million years, evade and subvert host immune responses, in part, by expression of immuno-modulatory molecules. Alphaherpesviruses express a single, broadly conserved chemokine decoy receptor, glycoprotein G (gG), which can bind multiple chemokine classes from multiple species, including human and mouse. Previously, we demonstrated that infection of chickens with an infectious laryngotracheitis virus (ILTV) mutant deficient in gG resulted in altered host immune responses compared to infection with wild-type virus. The ability of gG to disrupt the chemokine network has the potential to be used therapeutically. Here we investigated whether gG from ILTV or equine herpesvirus 1 (EHV-1) could modulate the protective immune response induced by the Helicobacter pylori vaccine antigen, catalase (KatA). Subcutaneous immunisation of mice with KatA together with EHV-1 gG, but not ILTV gG, induced significantly higher anti-KatA IgG than KatA alone. Importantly, subcutaneous or intranasal immunisation with KatA and EHV-1 gG both resulted in significantly lower colonization levels of H. pylori colonization following challenge, compared to mice vaccinated with KatA alone. Indeed, the lowest colonization levels were observed in mice vaccinated with KatA and EHV-1 gG, subcutaneously. In contrast, formulations containing ILTV gG did not affect H. pylori colonisation levels. The difference in efficacy between EHV-1 gG and ILTV gG may reflect the different spectrum of chemokines bound by the two proteins. Together, these data indicate that the immuno-modulatory properties of viral gGs could be harnessed for improving immune responses to vaccine antigens. Future studies should focus on the mechanism of action and whether gG may have other therapeutic applications.


Asunto(s)
Vacunas Bacterianas/inmunología , Glicoproteínas/inmunología , Infecciones por Helicobacter/prevención & control , Helicobacter pylori/inmunología , Herpesvirus Équido 1/inmunología , Herpesvirus Gallináceo 1/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Vacunas Bacterianas/administración & dosificación , Catalasa/administración & dosificación , Catalasa/inmunología , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Glicoproteínas/administración & dosificación , Infecciones por Helicobacter/inmunología , Helicobacter pylori/enzimología , Humanos , Inmunidad , Inmunización , Ratones , Ratones Endogámicos BALB C , Proteínas del Envoltorio Viral/administración & dosificación
13.
Trends Microbiol ; 21(5): 253-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23528348

RESUMEN

Helicobacter pylori can persist in the stomach of infected individuals for life, in the face of chronic inflammation and low pH. Efforts to develop vaccines have largely failed and, in the wake of emerging antibiotic resistance, novel therapeutic approaches must be considered. This review will discuss recent salient findings of host factors that modulate inflammatory responses to H. pylori with the aim of harnessing this knowledge for developing novel therapeutics. In addition, new approaches to vaccine development will be reviewed. Ultimately, the development of efficacious therapeutic interventions will likely need to consider host-pathogen interactions to enhance host immunity and circumvent bacterial evasion strategies.


Asunto(s)
Infecciones por Helicobacter/microbiología , Helicobacter pylori/fisiología , Interacciones Huésped-Patógeno , Animales , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/terapia , Helicobacter pylori/genética , Helicobacter pylori/inmunología , Humanos
14.
Vaccine ; 30(50): 7214-20, 2012 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-23084846

RESUMEN

Helicobacter pylori is an important pathogen of the human stomach, and the development of a protective vaccine has been an enticing goal for many years. The H. pylori antioxidant enzymes superoxide dismutase (SOD) and catalase (KatA) have been shown to be protective as vaccine antigens in mice, demonstrating that the organism's antioxidant enzyme system is a fruitful target for vaccine development. The research described here demonstrates that an additional antioxidant enzyme, thiolperoxidase (Tpx), is effective as a prophylactic vaccine antigen via both systemic and mucosal routes. The functional relationship between SOD, KatA and Tpx also provided an opportunity to investigate synergistic or additive effects when the three antigens were used in combination. Although the antigens still provided equivalent protection when administered in combination, no additional protection was observed. Moreover a decrease in antibody titres to the individual antigens was observed when delivered in combination via the nasal route, though not when injected subcutaneously. The findings of this paper demonstrate that the antioxidant system of H. pylori presents a particularly rich resource for vaccine development.


Asunto(s)
Antígenos Bacterianos/inmunología , Vacunas Bacterianas/inmunología , Infecciones por Helicobacter/prevención & control , Helicobacter pylori/enzimología , Helicobacter pylori/inmunología , Peroxidasa/inmunología , Administración Intranasal , Animales , Anticuerpos Antibacterianos/sangre , Vacunas Bacterianas/administración & dosificación , Femenino , Inyecciones Subcutáneas , Ratones , Ratones Endogámicos C57BL
15.
Am J Physiol Gastrointest Liver Physiol ; 302(6): G579-87, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22194421

RESUMEN

Helicobacter pylori is a microaerophilic, gram-negative pathogen of the human stomach. Despite the chronic active gastritis that develops following colonization, H. pylori is able to persist unharmed in the stomach for decades. Much of the damage caused by gastric inflammation results from the accumulation of reactive oxygen/nitrogen species within the stomach environment, which can induce oxidative damage in a wide range of biological molecules. Without appropriate defenses, this oxidative damage would be able to rapidly kill nearby H. pylori, but the organism employs a range of measures, including antioxidant enzymes, biological repair systems, and inhibitors of oxidant generation, to counter the attack. Despite the variety of measures employed to defend against oxidative injury, these processes are intimately interdependent, and any deficiency within the antioxidant system is generally sufficient to cause substantial impairment of H. pylori viability and persistence. This review provides an overview of the development of oxidative stress during H. pylori gastritis and examines the methods the organism uses to survive the resultant damage.


Asunto(s)
Helicobacter pylori/fisiología , Estrés Oxidativo/fisiología , Antioxidantes/metabolismo , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiología , Gastritis/metabolismo , Gastritis/microbiología , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/microbiología , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo
16.
Infect Immun ; 79(10): 4186-92, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21807907

RESUMEN

While gastric adenocarcinoma is the most serious consequence of Helicobacter pylori infection, not all infected persons develop this pathology. Individuals most at risk of this cancer are those in whom the bacteria colonize the acid-secreting region of the stomach and subsequently develop severe inflammation in the gastric corpus. It has been reported anecdotally that male mice become infected with greater numbers of H. pylori bacteria than female mice. While investigating this phenomenon, we found that increased H. pylori infection densities in male mice were not related to antibody production, and this phenomenon was not normalized by gonadectomy. However, the gastric pH in male 129/Sv mice was significantly elevated compared with that in female mice. Differences in colonization were evident within 1 day postinfection and significantly arose due to colonization of the gastric corpus region in male mice. This provided a potential model for comparing the effect of corpus colonization on the development of gastritis. This was explored using two models of H. pylori-induced inflammation, namely, 2-month infections of Muc1(-/-) mice and 6-month infections of wild-type 129/Sv mice. While H. pylori infection of female mice induced a severe, corpus-predominant atrophic gastritis, to our surprise, male mice developed minimal inflammation despite being colonized with significantly more H. pylori bacteria than female controls. Thus, colonization of the gastric corpus in male mice was associated with a loss of inflammation in that region. The suppression of inflammation concomitant with infection of the gastric corpus in male mice demonstrates a powerful localized suppression of inflammation induced at sites of H. pylori colonization.


Asunto(s)
Mucosa Gástrica/patología , Gastritis Atrófica/patología , Infecciones por Helicobacter/complicaciones , Helicobacter pylori/patogenicidad , Inflamación/patología , Estómago/patología , Animales , Modelos Animales de Enfermedad , Femenino , Mucosa Gástrica/microbiología , Gastritis Atrófica/inmunología , Gastritis Atrófica/microbiología , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/patología , Helicobacter pylori/aislamiento & purificación , Humanos , Concentración de Iones de Hidrógeno , Inflamación/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Estómago/microbiología
17.
Vet Res ; 42: 26, 2011 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-21314909

RESUMEN

Since the discovery that Helicobacter pylori causes a range of pathologies in the stomachs of infected humans, it has become apparent that Helicobacters are found in a diverse range of animal species where they are frequently associated with disease. In 2003 and 2004, there were two outbreaks of increased mortality associated with gastric bleeding and weight-loss in a captive colony of the Australian marsupial, the Stripe-faced Dunnart (Sminthopsis macroura). The presence of gastric pathology led to an investigation of potential Helicobacter pathogenesis in these animals. Histological examination revealed the presence of gastritis, and PCR analysis confirmed the presence of Helicobacter infection in the stomachs of these marsupials. Surprisingly, sequencing of 16S rRNA from these bacteria identified the species as H. pylori and PCR confirmed the strain to be positive for the important pathogenesis factor, cagA. We therefore describe, for the first time, an apparent reverse zoonotic infection of Stripe-faced Dunnarts with H. pylori. Already prone to pathological effects of stress (as experienced during breeding season), concomitant H. pylori infection appears to be a possible essential but not sufficient co-factor in prototypic gastric bleeding and weight loss in these marsupials. The Stripe-faced Dunnart could represent a new model for investigating Helicobacter-driven gastric pathology. Infections from their human handlers, specifically of H. pylori, may be a potential risk to captive colonies of marsupials.


Asunto(s)
Brotes de Enfermedades/veterinaria , Infecciones por Helicobacter/veterinaria , Helicobacter/genética , Helicobacter/aislamiento & purificación , Marsupiales , Zoonosis/epidemiología , Animales , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Recuento de Colonia Microbiana/veterinaria , Femenino , Helicobacter/metabolismo , Infecciones por Helicobacter/epidemiología , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/mortalidad , Humanos , Masculino , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa/veterinaria , ARN Ribosómico 16S/genética , ARN Ribosómico 16S/metabolismo , Análisis de Secuencia de ADN/veterinaria , Ureasa/genética , Ureasa/metabolismo , Victoria , Zoonosis/microbiología , Zoonosis/mortalidad
18.
Vaccine ; 29(7): 1514-8, 2011 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-21172379

RESUMEN

Helicobacter pylori, the major cause of gastric cancer, have mechanisms that allow colonization of the inhospitable gastric mucosa, including enzymes such as superoxide dismutase (SOD) which protect against reactive oxygen species. As SOD is essential for in vivo colonization, we theorized it might constitute a viable vaccine target. H. pylori SOD was expressed in E. coli and a purified recombinant protein used to vaccinate mice, prior to live H. pylori challenge. Partial protective immunity was induced, similar to that commonly observed with other antigens tested previously. This suggests SOD may have utility in a combination vaccine comprising several protective antigens.


Asunto(s)
Antígenos Bacterianos/inmunología , Vacunas Bacterianas/inmunología , Infecciones por Helicobacter/prevención & control , Helicobacter pylori/enzimología , Superóxido Dismutasa/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/genética , Escherichia coli/genética , Escherichia coli/inmunología , Femenino , Infecciones por Helicobacter/inmunología , Helicobacter pylori/inmunología , Inmunidad Humoral , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Superóxido Dismutasa/genética
19.
PLoS Pathog ; 5(10): e1000617, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19816567

RESUMEN

The bacterium Helicobacter pylori can cause peptic ulcer disease, gastric adenocarcinoma and MALT lymphoma. The cell-surface mucin MUC1 is a large glycoprotein which is highly expressed on the mucosal surface and limits the density of H. pylori in a murine infection model. We now demonstrate that by using the BabA and SabA adhesins, H. pylori bind MUC1 isolated from human gastric cells and MUC1 shed into gastric juice. Both H. pylori carrying these adhesins, and beads coated with MUC1 antibodies, induced shedding of MUC1 from MKN7 human gastric epithelial cells, and shed MUC1 was found bound to H. pylori. Shedding of MUC1 from non-infected cells was not mediated by the known MUC1 sheddases ADAM17 and MMP-14. However, knockdown of MMP-14 partially affected MUC1 release early in infection, whereas ADAM17 had no effect. Thus, it is likely that shedding is mediated both by proteases and by disassociation of the non-covalent interaction between the alpha- and beta-subunits. H. pylori bound more readily to MUC1 depleted cells even when the bacteria lacked the BabA and SabA adhesins, showing that MUC1 inhibits attachment even when bacteria cannot bind to the mucin. Bacteria lacking both the BabA and SabA adhesins caused less apoptosis in MKN7 cells than wild-type bacteria, having a greater effect than deletion of the CagA pathogenicity gene. Deficiency of MUC1/Muc1 resulted in increased epithelial cell apoptosis, both in MKN7 cells in vitro, and in H. pylori infected mice. Thus, MUC1 protects the epithelium from non-MUC1 binding bacteria by inhibiting adhesion to the cell surface by steric hindrance, and from MUC1-binding bacteria by acting as a releasable decoy.


Asunto(s)
Infecciones por Helicobacter/prevención & control , Helicobacter pylori/patogenicidad , Mucina-1/fisiología , Animales , Adhesión Bacteriana/fisiología , Células Epiteliales/fisiología , Mucosa Gástrica/microbiología , Glicosilación , Humanos , Ratones , Unión Proteica , Subunidades de Proteína , Estómago/microbiología , Estómago/fisiología
20.
Infect Immun ; 77(7): 2962-70, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19380476

RESUMEN

As the majority of human pathogens infect via a mucosal surface, delivery of killed vaccines by mucosal routes could potentially improve protection against many such organisms. Our ability to develop effective killed mucosal vaccines is inhibited by a lack of adjuvants that are safe and effective in humans. The Ulex europaeus agglutinin I (UEA-I) lectin specifically binds M cells lining the murine gastrointestinal tract. We explored the potential for M-cell-targeted vaccination of whole, killed Helicobacter pylori, the main causative agent of peptic ulcer disease and gastric cancer, and Campylobacter jejuni, the most common cause of diarrhea. Oral delivery of UEA-I-agglutinated H. pylori or C. jejuni induced a significant increase in both serum and intestinal antibody levels. This elevated response (i) required the use of whole bacteria, as it did not occur with lysate; (ii) was not mediated by formation of particulate clumps, as agglutination with a lectin with a different glycan specificity had no effect; and (iii) was not due to lectin-mediated, nonspecific immunostimulatory activity, as UEA-I codelivery with nonagglutinated bacteria did not enhance the response. Vaccination with UEA-I-agglutinated, killed whole H. pylori induced a protective response against subsequent live challenge that was as effective as that induced by cholera toxin adjuvant. Moreover, vaccination against C. jejuni by this approach resulted in complete protection against challenge in almost all animals. We believe that this is the first demonstration that targeting of whole killed bacteria to mucosal M cells can induce protective immunity without the addition of an immunostimulatory adjuvant.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Vacunas Bacterianas/inmunología , Infecciones por Campylobacter/prevención & control , Infecciones por Helicobacter/prevención & control , Lectinas de Plantas/farmacología , Adyuvantes Inmunológicos/administración & dosificación , Administración Oral , Animales , Anticuerpos Antibacterianos/análisis , Anticuerpos Antibacterianos/sangre , Vacunas Bacterianas/administración & dosificación , Infecciones por Campylobacter/patología , Recuento de Colonia Microbiana , Femenino , Infecciones por Helicobacter/patología , Inmunoglobulina A/análisis , Inmunoglobulina A/sangre , Inmunoglobulina G/análisis , Inmunoglobulina G/sangre , Mucosa Intestinal/química , Mucosa Intestinal/inmunología , Ratones , Ratones Endogámicos BALB C , Lectinas de Plantas/administración & dosificación , Índice de Severidad de la Enfermedad , Estómago/microbiología , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...