Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Bioinformatics ; 39(6)2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37279738

RESUMEN

SUMMARY: Adaptive Immune Receptor Repertoire Sequencing is a rapidly developing field that has advanced understanding of the role of the adaptive immune system in health and disease. Numerous tools have been developed to analyse the complex data produced by this technique but work to compare their accuracy and reliability has been limited. Thorough, systematic assessment of their performance is dependent on the ability to produce high quality simulated datasets with known ground truth. We have developed AIRRSHIP, a flexible and fast Python package that produces synthetic human B cell receptor sequences. AIRRSHIP uses a comprehensive set of reference data to replicate key mechanisms in the immunoglobulin recombination process, with a particular focus on junctional complexity. Repertoires generated by AIRRSHIP are highly similar to published data and all steps in the sequence generation process are recorded. These data can be used to not only determine the accuracy of repertoire analysis tools but can also, by tuning of the large number of user-controllable parameters, give insight into factors that contribute to inaccuracies in results. AVAILABILITY AND IMPLEMENTATION: AIRRSHIP is implemented in Python. It is available via https://github.com/Cowanlab/airrship and on PyPI at https://pypi.org/project/airrship/. Documentation can be found at https://airrship.readthedocs.io/.


Asunto(s)
Documentación , Programas Informáticos , Humanos , Reproducibilidad de los Resultados
2.
J Immunol ; 197(2): 429-40, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27288531

RESUMEN

The cytoplasmic phosphatase, protein tyrosine phosphatase nonreceptor type 22 (PTPN22), is a negative regulator of T cell signaling. Genome-wide association studies have shown that single-nucleotide polymorphisms in PTPN22 confer an increased risk of developing multiple autoimmune diseases in humans. The precise function of PTPN22 and how the variant protein contributes to autoimmunity is not well understood. To address this issue, we investigated the effect of PTPN22 deficiency on disease susceptibility in a mouse model of autoimmune arthritis. The SKG mouse expresses a hypomorphic mutant allele of ZAP70, which, upon exposure to fungal Ags, predisposes the mice to a CD4(+) T cell-mediated autoimmune arthritis that closely resembles rheumatoid arthritis in humans. Surprisingly, SKG Ptpn22(-/-) mice developed less severe mannan-induced arthritis compared with SKG mice. Diminution of disease was not due to significant alterations in thymocyte development or repertoire selection in SKG Ptpn22(-/-) mice, even though T cell-mediated signal transduction was improved. Instead, Ptpn22 deficiency appeared to bias CD4 Th cell differentiation away from the Th17 lineage, which is pathogenic in this setting, to a more Th1/T regulatory-focused response. These data show that even small perturbations in TCR signal transduction pathways can have profound consequences on the differentiation of T cell lineages and thus for the development of autoimmune diseases.


Asunto(s)
Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Linfocitos T CD4-Positivos/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 22/inmunología , Animales , Western Blotting , Diferenciación Celular/inmunología , Citometría de Flujo , Mananos/toxicidad , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Mutantes , Reacción en Cadena de la Polimerasa , Proteína Tirosina Fosfatasa no Receptora Tipo 22/deficiencia , Receptores de Antígenos de Linfocitos T/inmunología , Células Th17/inmunología
3.
PLoS Pathog ; 11(7): e1005022, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26134405

RESUMEN

Rosetting, the adhesion of Plasmodium falciparum-infected erythrocytes to uninfected erythrocytes, involves clonal variants of the parasite protein P. falciparum erythrocyte membrane protein 1 (PfEMP1) and soluble serum factors. While rosetting is a well-known phenotypic marker of parasites associated with severe malaria, the reason for this association remains unclear, as do the molecular details of the interaction between the infected erythrocyte (IE) and the adhering erythrocytes. Here, we identify for the first time a single serum factor, the abundant serum protease inhibitor α2-macroglobulin (α2M), which is both required and sufficient for rosetting mediated by the PfEMP1 protein HB3VAR06 and some other rosette-mediating PfEMP1 proteins. We map the α2M binding site to the C terminal end of HB3VAR06, and demonstrate that α2M can bind at least four HB3VAR06 proteins, plausibly augmenting their combined avidity for host receptors. IgM has previously been identified as a rosette-facilitating soluble factor that acts in a similar way, but it cannot induce rosetting on its own. This is in contrast to α2M and probably due to the more limited cross-linking potential of IgM. Nevertheless, we show that IgM works synergistically with α2M and markedly lowers the concentration of α2M required for rosetting. Finally, HB3VAR06+ IEs share the capacity to bind α2M with subsets of genotypically distinct P. falciparum isolates forming rosettes in vitro and of patient parasite isolates ex vivo. Together, our results are evidence that P. falciparum parasites exploit α2M (and IgM) to expand the repertoire of host receptors available for PfEMP1-mediated IE adhesion, such as the erythrocyte carbohydrate moieties that lead to formation of rosettes. It is likely that this mechanism also affects IE adhesion to receptors on vascular endothelium. The study opens opportunities for broad-ranging immunological interventions targeting the α2M--(and IgM-) binding domains of PfEMP1, which would be independent of the host receptor specificity of clinically important PfEMP1 antigens.


Asunto(s)
Eritrocitos/parasitología , Malaria Falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Formación de Roseta , alfa-Macroglobulinas/metabolismo , Animales , Humanos , Plasmodium falciparum/metabolismo
4.
Front Immunol ; 15: 1412211, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39011042

RESUMEN

Adoptive immunotherapy with Epstein-Barr virus (EBV)-specific T cells is an effective treatment for relapsed or refractory EBV-induced post-transplant lymphoproliferative disorders (PTLD) with overall survival rates of up to 69%. EBV-specific T cells have been conventionally made by repeated stimulation with EBV-transformed lymphoblastoid cell lines (LCL), which act as antigen-presenting cells. However, this process is expensive, takes many months, and has practical risks associated with live virus. We have developed a peptide-based, virus-free, serum-free closed system to manufacture a bank of virus-specific T cells (VST) for clinical use. We compared these with standard LCL-derived VST using comprehensive characterization and potency assays to determine differences that might influence clinical benefits. Multi-parameter flow cytometry revealed that peptide-derived VST had an expanded central memory population and less exhaustion marker expression than LCL-derived VST. A quantitative HLA-matched allogeneic cytotoxicity assay demonstrated similar specific killing of EBV-infected targets, though peptide-derived EBV T cells had a significantly higher expression of antiviral cytokines and degranulation markers after antigen recall. High-throughput T cell receptor-beta (TCRß) sequencing demonstrated oligoclonal repertoires, with more matches to known EBV-binding complementary determining region 3 (CDR3) sequences in peptide-derived EBV T cells. Peptide-derived products showed broader and enhanced specificities to EBV nuclear antigens (EBNAs) in both CD8 and CD4 compartments, which may improve the targeting of highly expressed latency antigens in PTLD. Importantly, peptide-based isolation and expansion allows rapid manufacture and significantly increased product yield over conventional LCL-based approaches.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Herpesvirus Humano 4 , Inmunoterapia Adoptiva , Péptidos , Humanos , Herpesvirus Humano 4/inmunología , Inmunoterapia Adoptiva/métodos , Péptidos/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/terapia , Línea Celular Transformada , Activación de Linfocitos/inmunología , Linfocitos T/inmunología
5.
Front Immunol ; 14: 1241474, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37638026

RESUMEN

B cells are key pathogenic drivers of chronic inflammation in rheumatoid arthritis (RA). There is limited understanding of the relationship between synovial B cell subsets and pathogenic antibody secreting cells (ASCs). This knowledge is crucial for the development of more targeted B-cell depleting therapies. While CD11c+ double-negative 2 (DN2) B cells have been suggested as an ASC precursor in lupus, to date there is no proven link between the two subsets in RA. We have used both single-cell gene expression and BCR sequencing to study synovial B cells from patients with established RA, in addition to flow cytometry of circulating B cells. To better understand the differentiation patterns within the diseased tissue, a combination of RNA-based trajectory inference and clonal lineage analysis of BCR relationships were used. Both forms of analysis indicated that DN2 B cells serve as a major precursors to synovial ASCs. This study advances our understanding of B cells in RA and reveals the origin of pathogenic ASCs in the RA synovium. Given the significant role of DN2 B cells as a progenitor to pathogenic B cells in RA, it is important to conduct additional research to investigate the origins of DN2 B cells in RA and explore their potential as therapeutic targets in place of the less specific pan-B cells depletion therapies currently in use.


Asunto(s)
Artritis Reumatoide , Subgrupos de Linfocitos B , Humanos , Células Plasmáticas , Linfocitos B , Células Productoras de Anticuerpos
6.
Parasitology ; 138(12): 1607-19, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21729355

RESUMEN

With the current paucity of vaccine targets for parasitic diseases, particularly those in childhood, the aim of this study was to compare protein expression and immune cross-reactivity between the trematodes Schistosoma haematobium, S. bovis and Echinostoma caproni in the hope of identifying novel intervention targets. Native adult parasite proteins were separated by 2-dimensional gel electrophoresis and identified through electrospray ionisation tandem mass spectrometry to produce a reference gel. Proteins from differential gel electrophoresis analyses of the three parasite proteomes were compared and screened against sera from hamsters infected with S. haematobium and E. caproni following 2-dimensional Western blotting. Differential protein expression between the three species was observed with circa 5% of proteins from S. haematobium showing expression up-regulation compared to the other two species. There was 91% similarity between the proteomes of the two Schistosoma species and 81% and 78·6% similarity between S. haematobium and S. bovis versus E. caproni, respectively. Although there were some common cross-species antigens, species-species targets were revealed which, despite evolutionary homology, could be due to phenotypic plasticity arising from different host-parasite relationships. Nevertheless, this approach helps to identify novel intervention targets which could be used as broad-spectrum candidates for future use in human and veterinary vaccines.


Asunto(s)
Echinostoma/inmunología , Equinostomiasis/parasitología , Proteínas del Helminto/análisis , Schistosoma haematobium/inmunología , Schistosoma/inmunología , Esquistosomiasis/parasitología , Animales , Antígenos Helmínticos/inmunología , Biomphalaria , Bulinus , Niño , Cricetinae , Reacciones Cruzadas , Echinostoma/metabolismo , Equinostomiasis/inmunología , Electroforesis en Gel Bidimensional , Proteínas del Helminto/inmunología , Proteínas del Helminto/metabolismo , Interacciones Huésped-Parásitos , Humanos , Masculino , Mesocricetus , Fenotipo , Proteoma , Proteómica , Schistosoma/metabolismo , Schistosoma haematobium/metabolismo , Esquistosomiasis/inmunología , Especificidad de la Especie , Regulación hacia Arriba
7.
Parasitology ; 138(12): 1519-33, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21813042

RESUMEN

Despite the overlapping distribution of Schistosoma haematobium and Plasmodium falciparum infections, few studies have investigated early immune responses to both parasites in young children resident in areas co-endemic for the parasites. This study measures infection levels of both parasites and relates them to exposure and immune responses in young children. Levels of IgM, IgE, IgG4 directed against schistosome cercariae, egg and adult worm and IgM, IgG directed against P. falciparum schizonts and the merozoite surface proteins 1 and 2 together with the cytokines IFN-γ, IL-4, IL-5, IL-10 and TNF-α were measured by ELISA in 95 Zimbabwean children aged 1-5 years. Schistosome infection prevalence was 14·7% and that of Plasmodium infection was 0% in the children. 43. 4% of the children showed immunological evidence of exposure to schistosome parasites and 13% showed immunological evidence of exposure to Plasmodium parasites. Schistosome-specific responses, indicative of exposure to parasite antigens, were positively associated with cercariae-specific IgE responses, while Plasmodium-specific responses, indicative of exposure to parasite antigens, were negatively associated with responses associated with protective immunity against Plasmodium. There was no significant association between schistosome-specific and Plasmodium-specific responses. Systemic cytokine levels rose with age as well as with schistosome infection and exposure. Overall the results show that (1) significantly more children are exposed to schistosome and Plasmodium infection than those currently infected and; (2) the development of protective acquired immunity commences in early childhood, although its effects on infection levels and pathology may take many years to become apparent.


Asunto(s)
Citocinas/análisis , Inmunoglobulinas/biosíntesis , Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Schistosoma haematobium/inmunología , Esquistosomiasis Urinaria/inmunología , Factores de Edad , Animales , Anticuerpos Antihelmínticos/biosíntesis , Anticuerpos Antihelmínticos/sangre , Anticuerpos Antiprotozoarios/biosíntesis , Anticuerpos Antiprotozoarios/sangre , Preescolar , Coinfección , Enfermedades Endémicas , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunoglobulinas/sangre , Lactante , Malaria Falciparum/epidemiología , Masculino , Prevalencia , Esquistosomiasis Urinaria/epidemiología , Zimbabwe/epidemiología
8.
Front Immunol ; 11: 587756, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33329568

RESUMEN

CD4+ αß T-cells are key mediators of the immune response to a first Plasmodium infection, undergoing extensive activation and splenic expansion during the acute phase of an infection. However, the clonality and clonal composition of this expansion has not previously been described. Using a comparative infection model, we sequenced the splenic CD4+ T-cell receptor repertoires generated over the time-course of a Plasmodium chabaudi infection. We show through repeat replicate experiments, single-cell RNA-seq, and analyses of independent RNA-seq data, that following a first infection - within a highly polyclonal expansion - T-effector repertoires are consistently dominated by TRBV3 gene usage. Clustering by sequence similarity, we find the same dominant clonal signature is expanded across replicates in the acute phase of an infection, revealing a conserved pathogen-specific T-cell response that is consistently a hallmark of a first infection, but not expanded upon re-challenge. Determining the host or parasite factors driving this conserved response may uncover novel immune targets for malaria therapeutic purposes.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Malaria/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Enfermedad Aguda , Animales , Femenino , Malaria/genética , Ratones Endogámicos C57BL , Plasmodium chabaudi , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Bazo/citología , Bazo/inmunología
9.
Front Immunol ; 11: 395, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32265907

RESUMEN

B cells are critical for promoting autoimmunity and the success of B cell depletion therapy in rheumatoid arthritis (RA) confirms their importance in driving chronic inflammation. Whilst disease specific autoantibodies are useful diagnostically, our understanding of the pathogenic B cell repertoire remains unclear. Defining it would lead to novel insights and curative treatments. To address this, we have undertaken the largest study to date of over 150 RA patients, utilizing next generation sequencing (NGS) to analyze up to 200,000 BCR sequences per patient. The full-length antigen-binding variable region of the heavy chain (IgGHV) of the IgG B cell receptor (BCR) were sequenced. Surprisingly, RA patients do not express particular clonal expansions of B cells at diagnosis. Rather they express a polyclonal IgG repertoire with a significant increase in BCRs that have barely mutated away from the germline sequence. This pattern remains even after commencing disease modifying therapy. These hypomutated BCRs are expressed by TNF-alpha secreting IgG+veCD27-ve B cells, that are expanded in RA peripheral blood and enriched in the rheumatoid synovium. A similar B cell repertoire is expressed by patients with Sjögren's syndrome. A rate limiting step in the initiation of autoimmunity is the activation of B cells and this data reveals that a sizeable component of the human autoimmune B cell repertoire consists of polyclonal, hypomutated IgG+ve B cells, that may play a critical role in driving chronic inflammation.


Asunto(s)
Artritis Reumatoide/inmunología , Autoinmunidad , Linfocitos B/inmunología , Genes de Inmunoglobulinas , Inmunoglobulina G/genética , Subgrupos Linfocitarios/inmunología , Antirreumáticos/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/patología , Autoanticuerpos/inmunología , Linaje de la Célula , Células Clonales , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunoglobulina G/análisis , Cadenas Pesadas de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/genética , Activación de Linfocitos , Receptores de Antígenos de Linfocitos B/genética , Síndrome de Sjögren/inmunología , Síndrome de Sjögren/patología , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/análisis , Factor de Necrosis Tumoral alfa/metabolismo
10.
Methods Mol Biol ; 1623: 281-301, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28589363

RESUMEN

Methods for tracking B-cell repertoires and clonal history in normal and malignant B-cells based on immunoglobulin variable region (IGV) gene analysis have developed rapidly with the advent of massive parallel next-generation sequencing (mpNGS) protocols. mpNGS permits a depth of analysis of IGV genes not hitherto feasible, and presents challenges of bioinformatics analysis, which can be readily met by current pipelines. This strategy offers a potential resolution of B-cell usage at a depth that may capture fully the natural state, in a given biological setting. Conventional methods based on RT-PCR amplification and Sanger sequencing are also available where mpNGS is not accessible. Each method offers distinct advantages. Conventional methods for IGV gene sequencing are readily adaptable to most laboratories and provide an ease of analysis to capture salient features of B-cell use. This chapter describes two methods in detail for analysis of IGV genes, mpNGS and conventional RT-PCR with Sanger sequencing.


Asunto(s)
Linfocitos B/metabolismo , Rastreo Celular/métodos , Evolución Clonal/genética , Genes de Inmunoglobulinas , Linfoma de Células B/genética , Linfocitos B/inmunología , Linfocitos B/patología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Evolución Clonal/inmunología , Biología Computacional/métodos , Centro Germinal/citología , Centro Germinal/inmunología , Centro Germinal/metabolismo , Centro Germinal/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Linfoma de Células B/inmunología , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Hipermutación Somática de Inmunoglobulina , Estadística como Asunto
11.
Front Immunol ; 8: 1952, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29403471

RESUMEN

The chronic autoimmune inflammatory diseases, systemic lupus erythematosus and Sjogren's syndrome, develop when tolerance to apoptotic cells (ACs) is lost. We have previously reported that this tolerance is maintained by innate-like, IL-10 secreting regulatory B cells. Two questions remained. First, do these regulatory B cells belong predominantly to a single subset of steady-state B cells and second, what is their specificity? We report here that innate-like B cells with markers characteristic for B1a cells (CD43+veCD19hiCD5+veIgMhiIgDlo) constitute 80% of splenic and 96% of peritoneal B cells that respond to ACs by secreting IL-10. AC responsive B1a cells secrete self-reactive natural antibodies (NAbs) and IL-10, which is augmented by toll-like receptor (TLR) 7 or TLR9 stimulation. In so doing, they both accelerate the clearance of dying cells by macrophages and inhibit their potential to mount proinflammatory immune responses. While B1a cells make prolonged contact with ACs, they do not require TIM1 or complement to mediate their regulatory function. In an animal model of neural inflammation (experimental autoimmune encephalomyelitis), just 105 activated B1a B cells was sufficient to restrain inflammation. Activated B1a B cells also induced antigen-specific T cells to secrete IL-10. Hence, regulatory B1a cells specifically recognize and augment tolerance to apoptotic self via IL-10 and NAbs; but once activated, can also prevent autoimmune mediated inflammation.

12.
Oncotarget ; 6(15): 13229-40, 2015 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-25929340

RESUMEN

Human multiple myeloma (MM) is characterized by accumulation of malignant terminally differentiated plasma cells (PCs) in the bone marrow (BM), raising the question when during maturation neoplastic transformation begins. Immunoglobulin IGHV genes carry imprints of clonal tumor history, delineating somatic hypermutation (SHM) events that generally occur in the germinal center (GC). Here, we examine MM-derived IGHV genes using massive parallel deep sequencing, comparing them with profiles in normal BM PCs. In 4/4 presentation IgG MM, monoclonal tumor-derived IGHV sequences revealed significant evidence for intraclonal variation (ICV) in mutation patterns. IGHV sequences of 2/2 normal PC IgG populations revealed dominant oligoclonal expansions, each expansion also displaying mutational ICV. Clonal expansions in MM and in normal BM PCs reveal common IGHV features. In such MM, the data fit a model of tumor origins in which neoplastic transformation is initiated in a GC B-cell committed to terminal differentiation but still targeted by on-going SHM. Strikingly, the data parallel IGHV clonal sequences in some monoclonal gammopathy of undetermined significance (MGUS) known to display on-going SHM imprints. Since MGUS generally precedes MM, these data suggest origins of MGUS and MM with IGHV gene mutational ICV from the same GC B-cell, arising via a distinctive pathway.


Asunto(s)
Genes de Inmunoglobulinas/genética , Centro Germinal/patología , Cadenas Pesadas de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/genética , Mieloma Múltiple/genética , Células de la Médula Ósea/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Gammopatía Monoclonal de Relevancia Indeterminada/patología , Mieloma Múltiple/patología , Mutación , Células Plasmáticas/patología , Hipermutación Somática de Inmunoglobulina/fisiología
13.
Neuromuscul Disord ; 25(2): 127-36, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25454731

RESUMEN

Emery-Dreifuss muscular dystrophy (EDMD) is a neuromuscular disease characterized by early contractures, slowly progressive muscular weakness and life-threatening cardiac arrhythmia that can develop into cardiomyopathy. In X-linked EDMD (EDMD1), female carriers are usually unaffected. Here we present a clinical description and in vitro characterization of a mildly affected EDMD1 female carrying the heterozygous EMD mutation c.174_175delTT; p.Y59* that yields loss of protein. Muscle tissue sections and cultured patient myoblasts exhibited a mixed population of emerin-positive and -negative cells; thus uneven X-inactivation was excluded as causative. Patient blood cells were predominantly emerin-positive, but considerable nuclear lobulation was observed in non-granulocyte cells - a novel phenotype in EDMD. Both emerin-positive and emerin-negative myoblasts exhibited spontaneous differentiation in tissue culture, though emerin-negative myoblasts were more proliferative than emerin-positive cells. The preferential proliferation of emerin-negative myoblasts together with the high rate of spontaneous differentiation in both populations suggests that loss of functional satellite cells might be one underlying mechanism for disease pathology. This could also account for the slowly developing muscle phenotype.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Distrofia Muscular de Emery-Dreifuss/patología , Mioblastos/patología , Adolescente , Adulto , Edad de Inicio , Antígenos CD , Autoantígenos/metabolismo , Cardiomiopatías/etiología , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/genética , Proliferación Celular/genética , Células Cultivadas , Niño , Salud de la Familia , Femenino , Citometría de Flujo , Humanos , Antígeno Ki-67/metabolismo , Lamina Tipo A/metabolismo , Imagen por Resonancia Magnética , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Distrofia Muscular de Emery-Dreifuss/complicaciones , Distrofia Muscular de Emery-Dreifuss/genética , Mioblastos/metabolismo , Proteínas Nucleares/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transfección , Adulto Joven
14.
PLoS One ; 9(1): e87198, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24489871

RESUMEN

Development of effective malaria vaccines is hampered by the problem of producing correctly folded Plasmodium proteins for use as vaccine components. We have investigated the use of a novel ciliate expression system, Tetrahymena thermophila, as a P. falciparum vaccine antigen platform. A synthetic vaccine antigen composed of N-terminal and C-terminal regions of merozoite surface protein-1 (MSP-1) was expressed in Tetrahymena thermophila. The recombinant antigen was secreted into the culture medium and purified by monoclonal antibody (mAb) affinity chromatography. The vaccine was immunogenic in MF1 mice, eliciting high antibody titers against both N- and C-terminal components. Sera from immunized animals reacted strongly with P. falciparum parasites from three antigenically different strains by immunofluorescence assays, confirming that the antibodies produced are able to recognize parasite antigens in their native form. Epitope mapping of serum reactivity with a peptide library derived from all three MSP-1 Block 2 serotypes confirmed that the MSP-1 Block 2 hybrid component of the vaccine had effectively targeted all three serotypes of this polymorphic region of MSP-1. This study has successfully demonstrated the use of Tetrahymena thermophila as a recombinant protein expression platform for the production of malaria vaccine antigens.


Asunto(s)
Vacunas contra la Malaria/biosíntesis , Malaria Falciparum/prevención & control , Proteína 1 de Superficie de Merozoito/biosíntesis , Tetrahymena thermophila/metabolismo , Vacunación , Animales , Animales no Consanguíneos , Anticuerpos Antiprotozoarios/sangre , Mapeo Epitopo , Femenino , Humanos , Vacunas contra la Malaria/inmunología , Malaria Falciparum/sangre , Malaria Falciparum/inmunología , Proteína 1 de Superficie de Merozoito/inmunología , Ratones , Plasmodium falciparum/inmunología , Potencia de la Vacuna
15.
PLoS One ; 9(1): e83704, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24421900

RESUMEN

The Block 2 region of the merozoite surface protein-1 (MSP-1) of Plasmodium falciparum has been identified as a target of protective immunity by a combination of seroepidemiology and parasite population genetics. Immunogenicity studies in small animals and Aotus monkeys were used to determine the efficacy of recombinant antigens derived from this region of MSP-1 as a potential vaccine antigen. Aotus lemurinus griseimembra monkeys were immunized three times with a recombinant antigen derived from the Block 2 region of MSP-1 of the monkey-adapted challenge strain, FVO of Plasmodium falciparum, using an adjuvant suitable for use in humans. Immunofluorescent antibody assays (IFA) against erythrocytes infected with P. falciparum using sera from the immunized monkeys showed that the MSP-1 Block 2 antigen induced significant antibody responses to whole malaria parasites. MSP-1 Block 2 antigen-specific enzyme-linked immunosorbent assays (ELISA) showed no significant differences in antibody titers between immunized animals. Immunized animals were challenged with the virulent P. falciparum FVO isolate and monitored for 21 days. Two out of four immunized animals were able to control their parasitaemia during the follow-up period, whereas two out of two controls developed fulminating parasitemia. Parasite-specific serum antibody titers measured by IFA were four-fold higher in protected animals than in unprotected animals. In addition, peptide-based epitope mapping of serum antibodies from immunized Aotus showed distinct differences in epitope specificities between protected and unprotected animals.


Asunto(s)
Formación de Anticuerpos/inmunología , Haplorrinos/inmunología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Malaria Falciparum/prevención & control , Proteína 1 de Superficie de Merozoito/inmunología , Plasmodium falciparum/inmunología , Adyuvantes Inmunológicos , Secuencia de Aminoácidos , Animales , Especificidad de Anticuerpos/inmunología , Antígenos de Protozoos/inmunología , Mapeo Epitopo , Epítopos/química , Epítopos/inmunología , Haplorrinos/sangre , Haplorrinos/parasitología , Humanos , Inmunización , Malaria Falciparum/sangre , Malaria Falciparum/parasitología , Datos de Secuencia Molecular , Parasitemia/inmunología , Parasitemia/parasitología , Proteínas Recombinantes/inmunología
16.
J Trop Med ; 2013: 283619, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23840222

RESUMEN

Urogenital schistosomiasis, due to Schistosoma haematobium, is endemic in sub-Saharan Africa. Control is by targeted treatment with praziquantel but preschool age children are excluded from control programs. Immunological studies on the effect of treatment at this young age are scarce. In light of studies in older individuals showing that praziquantel alters antischistosome immune responses and responses to bystander antigens, this study aims to investigate how these responses would be affected by treatment at this young age. Antibody responses directed against schistosome antigens, Plasmodium falciparum crude and recombinant antigens, and the allergen house dust mite were measured in children aged 3 to 5 years before and 6 weeks after treatment. The change in serological recognition of schistosome proteins was also investigated. Treatment augmented antischistosome IgM and IgE responses. The increase in IgE responses directed against adult worm antigens was accompanied by enhanced antigen recognition by sera from the children. Antibody responses directed against Plasmodium antigens were not significantly affected by praziquantel treatment nor were levels of allergen specific responses. Overall, praziquantel treatment enhanced, quantitatively and qualitatively, the antiworm responses associated with protective immunity but did not alter Plasmodium-specific responses or allergen-specific responses which mediate pathology in allergic disease.

17.
PLoS One ; 6(10): e26616, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22073118

RESUMEN

Polymorphic parasite antigens are known targets of protective immunity to malaria, but this antigenic variation poses challenges to vaccine development. A synthetic MSP-1 Block 2 construct, based on all polymorphic variants found in natural Plasmodium falciparum isolates has been designed, combined with the relatively conserved Block 1 sequence of MSP-1 and expressed in E.coli. The MSP-1 Hybrid antigen has been produced with high yield by fed-batch fermentation and purified without the aid of affinity tags resulting in a pure and extremely thermostable antigen preparation. MSP-1 hybrid is immunogenic in experimental animals using adjuvants suitable for human use, eliciting antibodies against epitopes from all three Block 2 serotypes. Human serum antibodies from Africans naturally exposed to malaria reacted to the MSP-1 hybrid as strongly as, or better than the same serum reactivities to individual MSP-1 Block 2 antigens, and these antibody responses showed clear associations with reduced incidence of malaria episodes. The MSP-1 hybrid is designed to induce a protective antibody response to the highly polymorphic Block 2 region of MSP-1, enhancing the repertoire of MSP-1 Block 2 antibody responses found among immune and semi-immune individuals in malaria endemic areas. The target population for such a vaccine is young children and vulnerable adults, to accelerate the acquisition of a full range of malaria protective antibodies against this polymorphic parasite antigen.


Asunto(s)
Anticuerpos Antiprotozoarios/sangre , Anticuerpos Antiprotozoarios/inmunología , Inmunoglobulina G/inmunología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Proteína 1 de Superficie de Merozoito/inmunología , Plasmodium falciparum/inmunología , Adolescente , Adulto , Animales , Anticuerpos Monoclonales/inmunología , Estudios de Casos y Controles , Niño , Preescolar , Estudios Transversales , Ensayo de Inmunoadsorción Enzimática , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Inmunización , Immunoblotting , Macaca mulatta , Malaria Falciparum/prevención & control , Ratones , Ratones Endogámicos DBA , Plasmodium falciparum/crecimiento & desarrollo , Conejos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
18.
Vaccine ; 28(18): 3231-7, 2010 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-20188176

RESUMEN

Induction of immunity at mucosal surfaces is thought to be an essential feature in the protection of the host against the many pathogens that gain access through these surfaces. Here we describe how strong local and systemic immune responses can be generated when proteins are genetically conjugated to pneumolysin (PLY) from Streptococcus pneumoniae. Using green fluorescent protein (eGFP) and PsaA from S. pneumoniae, we have shown that genetic fusion (eGFPPLY and PsaAPLY) is essential to ensure high levels of antigen specific IgG and IgA in the serum and at mucosal surfaces. This form of vaccination is highly effective with antigen specific antibodies detected after a single dose of nanogram quantities of the conjugated proteins. In addition, generation of a non-toxic variant (eGFPDelta6PLY) indicated that while the toxic activity of PLY was not essential for adjuvanticity, it contributed to the magnitude of the response generated. Whilst vaccination with the PsaAPLY fusion proteins did not protect the animals from challenge, these studies confirm the utility of pneumolysin to act as a novel mucosal adjuvant to substantially increase the local and systemic humoral response to genetically fused protein antigens.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Inmunidad Mucosa , Proteínas/inmunología , Estreptolisinas/farmacología , Vacunas/inmunología , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/inmunología , Adyuvantes Inmunológicos/genética , Adyuvantes Inmunológicos/toxicidad , Animales , Anticuerpos/sangre , Proteínas Bacterianas/genética , Proteínas Bacterianas/farmacología , Proteínas Bacterianas/toxicidad , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/inmunología , Humanos , Inmunoglobulina A/análisis , Inmunoglobulina A/sangre , Inmunoglobulina G/sangre , Lipoproteínas/genética , Lipoproteínas/inmunología , Ratones , Ratones Endogámicos BALB C , Proteínas/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Eliminación de Secuencia , Estreptolisinas/genética , Estreptolisinas/toxicidad , Vacunas/genética , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
19.
Mol Immunol ; 46(7): 1561-7, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19200600

RESUMEN

The unique species specificity of the bacterial cytolysin intermedilysin is explained by its requirement for the human complement regulator CD59 as the primary receptor. Binding studies using individual domains of intermedilysin mapped the CD59-binding site to domain 4 and swap mutants between human and rabbit (non-intermedilysin-binding) CD59 implicated a short sequence (residues 42-59) in human CD59 in binding intermedilysin. We set out to map more closely the CD59 binding site in intermedilysin. We first looked for regions of homology between domain 4 in intermedilysin and the terminal complement components that bind CD59, C8 and C9. A nine amino acid sequence immediately adjacent the undecapeptide segment in intermedilysin domain 4 matched (5 of 9 identical, 3 of 9 conserved) a sequence in C9. A peptide containing this sequence caused dose-dependent inhibition of intermedilysin-mediated lysis of human erythrocytes and rendered erythrocytes more susceptible to complement lysis. Surface plasmon resonance analysis of intermedilysin binding to immobilized CD59 revealed saturable fast-on, fast-off binding and a calculated affinity of 4.9 nM. Substitution of three residues from the putative binding site caused a 5-fold reduction in lytic potency of intermedilysin and reduced affinity for immobilized CD59 by 2.5-fold. The demonstration that a peptide modeled on the CD59-binding site inhibits intermedilysin-mediated haemolysis leads us to suggest that such peptides might be useful in treating infections caused by intermedilysin-producing bacteria.


Asunto(s)
Bacteriocinas/metabolismo , Antígenos CD59/metabolismo , Secuencia de Aminoácidos , Animales , Bacteriocinas/química , Sitios de Unión , Antígenos CD59/química , Células CHO , Cricetinae , Cricetulus , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Hemólisis/fisiología , Hemolíticos/química , Hemolíticos/metabolismo , Humanos , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Unión Proteica , Mapeo de Interacción de Proteínas , Homología de Secuencia de Aminoácido , Especificidad por Sustrato
20.
Vaccine ; 25(41): 7197-205, 2007 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-17765365

RESUMEN

Anthrolysin O (ALO) is a toxin produced by Bacillus anthracis, the causative agent of anthrax. It is a member of the cholesterol-dependent cytolysin (CDC) group of toxins, many of which are potential vaccine candidates that protect against their producing organisms. Pore formation by ALO was studied by transmission electron microscopy and pores were found to be consistent with those formed by other members of this toxin family. We constructed and characterised a novel genetic toxoid of anthrolysin O, Delta6mALO, which was able to bind to cells but was incapable of pore-formation or haemolysis. The capacity of the haemolytic and non-haemolytic forms of ALO to protect against challenge with the toxin or B. anthracis was determined. Immunisation with both active and non-haemolytic forms of ALO elicited protection against lethal i.v. challenge with ALO but neither was protective against B. anthracis in a murine i.p. challenge model. Immunisation with another CDC, pneumolysin, did not confer cross-protection against challenge with ALO. Histopathological investigation following lethal i.v. challenge with ALO revealed acute pathology in the lungs with occlusion of alveolar vessels by fibrin deposits.


Asunto(s)
Vacunas contra el Carbunco/inmunología , Carbunco/prevención & control , Antígenos Bacterianos/toxicidad , Bacillus anthracis/inmunología , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/toxicidad , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/toxicidad , Intoxicación/prevención & control , Toxoides/inmunología , Animales , Carbunco/inmunología , Vacunas contra el Carbunco/genética , Vacunas contra el Carbunco/toxicidad , Antitoxinas/sangre , Proteínas Bacterianas/genética , Eritrocitos/efectos de los fármacos , Eritrocitos/ultraestructura , Femenino , Hemólisis , Humanos , Inmunoglobulina G/sangre , Pulmón/patología , Glicoproteínas de Membrana/genética , Ratones , Microscopía Electrónica de Transmisión , Intoxicación/inmunología , Análisis de Supervivencia , Toxoides/genética , Toxoides/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA