Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Respir Crit Care Med ; 196(12): 1526-1534, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-28915062

RESUMEN

RATIONALE: Recombinant fragment of human surfactant protein D (rfhSP-D) has been shown to suppress house dust mite- and Aspergillus fumigatus-induced allergic inflammation in murine models. OBJECTIVES: We sought to elucidate the effect of rfhSP-D on high-affinity IgE receptor- and CD23-mediated, grass pollen-induced allergic inflammatory responses. METHODS: rfhSP-D, containing homotrimeric neck and lectin domains, was expressed in Escherichia coli BL21(λDE3)pLysS cells. Peripheral blood mononuclear cells and sera were obtained from individuals with grass pollen allergy (n = 27). The effect of rfhSP-D on basophil activation and histamine release was measured by flow cytometry. IgE-facilitated allergen binding and presentation were assessed by flow cytometry. T-helper cell type 2 (Th2) cytokines were measured in cell culture supernatants. The effect of rfhSP-D on IgE production by B cells when stimulated with CD40L, IL-4, and IL-21 was also determined. MEASUREMENTS AND MAIN RESULTS: rfhSP-D bound to Phleum pratense in a dose- and calcium-dependent manner. Allergen-induced basophil responsiveness and histamine release were inhibited in the presence of rfhSP-D, as measured by CD63, CD203c (P = 0.0086, P = 0.04205), and intracellularly labeled diamine oxidase (P = 0.0003, P = 0.0148). The binding of allergen-IgE complexes to B cells was reduced by 51% (P = 0.002) in the presence of rfhSP-D. This decrease was concomitant with reduction in CD23 expression on B cells (P < 0.001). rfhSP-D suppressed allergen-driven CD27-CD4+CRTh2+ T-cell proliferation (P < 0.01), IL-4, and IL-5 levels (all P < 0.01). Moreover, rfhSP-D inhibited CD40L/IL-4- and IL-21-mediated IgE production (77.12%; P = 0.02) by B cells. CONCLUSIONS: For the first time, to our knowledge, we show that rfhSP-D inhibited allergen-induced basophil responses at a single-cell level and suppressed CD23-mediated facilitated allergen presentation and Th2 cytokine production. In addition, rfhSP-D inhibited IgE synthesis by B cells, which is also a novel observation.


Asunto(s)
Linfocitos B/inmunología , Basófilos/inmunología , Hipersensibilidad/inmunología , Inflamación/inmunología , Poaceae/inmunología , Polen/inmunología , Proteína D Asociada a Surfactante Pulmonar/inmunología , Adulto , Alérgenos/sangre , Alérgenos/inmunología , Femenino , Citometría de Flujo , Humanos , Hipersensibilidad/sangre , Hipersensibilidad/prevención & control , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Inflamación/sangre , Inflamación/prevención & control , Masculino , Persona de Mediana Edad , Proteína D Asociada a Surfactante Pulmonar/sangre , Receptores de IgE/sangre , Receptores de IgE/inmunología , Células Th2 , Adulto Joven
2.
BMC Infect Dis ; 14: 660, 2014 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-25466778

RESUMEN

BACKGROUND: There is an urgent need for improved vaccines to protect against tuberculosis. The currently available vaccine Bacille Calmette-Guerin (BCG) has varying immunogenicity and efficacy across different populations for reasons not clearly understood. MVA85A is a modified vaccinia virus expressing antigen 85A from Mycobacterium tuberculosis which has been in clinical development since 2002 as a candidate vaccine to boost BCG-induced protection. A recent efficacy trial in South African infants failed to demonstrate enhancement of protection over BCG alone. The immunogenicity was lower than that seen in UK trials. The enzyme Indoleamine 2,3-dioxygenase (IDO) catalyses the first and rate-limiting step in the breakdown of the essential amino acid tryptophan. T cells are dependent on tryptophan and IDO activity suppresses T-cell proliferation and function. METHODS: Using samples collected during phase I trials with MVA85A across the UK and South Africa we have investigated the relationship between vaccine immunogenicity and IDO using IFN-γ ELISPOT, qPCR and liquid chromatography mass spectrometry. RESULTS: We demonstrate an IFN-γ dependent increase in IDO mRNA expression in peripheral blood mononuclear cells (PBMC) following MVA85A vaccination in UK subjects. IDO mRNA correlates positively with the IFN-γ ELISPOT response indicating that vaccine specific induction of IDO in PBMC is unlikely to limit the development of vaccine specific immunity. IDO activity in the serum of volunteers from the UK and South Africa was also assessed. There was no change in serum IDO activity following MVA85A vaccination. However, we observed higher baseline IDO activity in South African volunteers when compared to UK volunteers. In both UK and South African serum samples, baseline IDO activity negatively correlated with vaccine-specific IFN-γ responses, suggesting that IDO activity may impair the generation of a CD4+ T cell memory response. CONCLUSIONS: Baseline IDO activity was higher in South African volunteers when compared to UK volunteers, which may represent a potential mechanism for the observed variation in vaccine immunogenicity in South African and UK populations and may have important implications for future vaccination strategies. TRIAL REGISTRATION: Trials are registered at ClinicalTrials.gov; UK cohort NCT00427830, UK LTBI cohort NCT00456183, South African cohort NCT00460590, South African LTBI cohort NCT00480558.


Asunto(s)
Indolamina-Pirrol 2,3,-Dioxigenasa/efectos de los fármacos , Interferón gamma/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Activación de Linfocitos/efectos de los fármacos , Mycobacterium tuberculosis/inmunología , ARN Mensajero/metabolismo , Vacunas contra la Tuberculosis/farmacología , Tuberculosis/prevención & control , Adulto , Vacuna BCG , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Ensayo de Immunospot Ligado a Enzimas , Femenino , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/sangre , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Leucocitos Mononucleares/metabolismo , Activación de Linfocitos/inmunología , Masculino , Persona de Mediana Edad , Sudáfrica , Reino Unido , Vacunación , Vacunas de ADN , Adulto Joven
3.
Front Immunol ; 15: 1352022, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38698856

RESUMEN

The complement system is an innate immune mechanism against microbial infections. It involves a cascade of effector molecules that is activated via classical, lectin and alternative pathways. Consequently, many pathogens bind to or incorporate in their structures host negative regulators of the complement pathways as an evasion mechanism. Factor H (FH) is a negative regulator of the complement alternative pathway that protects "self" cells of the host from non-specific complement attack. FH has been shown to bind viruses including human influenza A viruses (IAVs). In addition to its involvement in the regulation of complement activation, FH has also been shown to perform a range of functions on its own including its direct interaction with pathogens. Here, we show that human FH can bind directly to IAVs of both human and avian origin, and the interaction is mediated via the IAV surface glycoprotein haemagglutinin (HA). HA bound to common pathogen binding footprints on the FH structure, complement control protein modules, CCP 5-7 and CCP 15-20. The FH binding to H1 and H3 showed that the interaction overlapped with the receptor binding site of both HAs, but the footprint was more extensive for the H3 HA than the H1 HA. The HA - FH interaction impeded the initial entry of H1N1 and H3N2 IAV strains but its impact on viral multicycle replication in human lung cells was strain-specific. The H3N2 virus binding to cells was significantly inhibited by preincubation with FH, whereas there was no alteration in replicative rate and progeny virus release for human H1N1, or avian H9N2 and H5N3 IAV strains. We have mapped the interaction between FH and IAV, the in vivo significance of which for the virus or host is yet to be elucidated.


Asunto(s)
Factor H de Complemento , Glicoproteínas Hemaglutininas del Virus de la Influenza , Virus de la Influenza A , Gripe Humana , Unión Proteica , Humanos , Factor H de Complemento/metabolismo , Factor H de Complemento/inmunología , Animales , Gripe Humana/inmunología , Gripe Humana/virología , Gripe Humana/metabolismo , Virus de la Influenza A/inmunología , Virus de la Influenza A/fisiología , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Sitios de Unión , Gripe Aviar/virología , Gripe Aviar/inmunología , Gripe Aviar/metabolismo , Aves/virología , Interacciones Huésped-Patógeno/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Subtipo H9N2 del Virus de la Influenza A/inmunología
4.
Nat Med ; 10(11): 1240-4, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15502839

RESUMEN

Protective immunity against Mycobacterium tuberculosis depends on the generation of a T(H)1-type cellular immune response, characterized by the secretion of interferon-gamma (IFN-gamma) from antigen-specific T cells. The induction of potent cellular immune responses by vaccination in humans has proven difficult. Recombinant viral vectors, especially poxviruses and adenoviruses, are particularly effective at boosting previously primed CD4(+) and CD8(+) T-cell responses against a number of intracellular pathogens in animal studies. In the first phase 1 study of any candidate subunit vaccine against tuberculosis, recombinant modified vaccinia virus Ankara (MVA) expressing antigen 85A (MVA85A) was found to induce high levels of antigen-specific IFN-gamma-secreting T cells when used alone in bacille Calmette-Guerin (BCG)-naive healthy volunteers. In volunteers who had been vaccinated 0.5-38 years previously with BCG, substantially higher levels of antigen-specific IFN-gamma-secreting T cells were induced, and at 24 weeks after vaccination these levels were 5-30 times greater than in vaccinees administered a single BCG vaccination. Boosting vaccinations with MVA85A could offer a practical and efficient strategy for enhancing and prolonging antimycobacterial immunity in tuberculosis-endemic areas.


Asunto(s)
Aciltransferasas/inmunología , Antígenos Bacterianos/inmunología , Mycobacterium tuberculosis/inmunología , Vacunas contra la Tuberculosis/inmunología , Tuberculosis/prevención & control , Virus Vaccinia/inmunología , Adulto , Ensayo de Inmunoadsorción Enzimática/métodos , Mapeo Epitopo , Humanos , Inmunidad Celular/inmunología , Interferón gamma/inmunología , Persona de Mediana Edad , Linfocitos T Colaboradores-Inductores/inmunología , Tuberculosis/inmunología , Vacunas Sintéticas/inmunología , Virus Vaccinia/metabolismo
5.
Am J Respir Crit Care Med ; 179(8): 724-33, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19151191

RESUMEN

RATIONALE: An effective new tuberculosis (TB) vaccine regimen must be safe in individuals with latent TB infection (LTBI) and is a priority for global health care. OBJECTIVES: To evaluate the safety and immunogenicity of a leading new TB vaccine, recombinant Modified Vaccinia Ankara expressing Antigen 85A (MVA85A) in individuals with LTBI. METHODS: An open-label, phase I trial of MVA85A was performed in 12 subjects with LTBI recruited from TB contact clinics in Oxford and London or by poster advertisements in Oxford hospitals. Patients were assessed clinically and had blood samples drawn for immunological analysis over a 52-week period after vaccination with MVA85A. Thoracic computed tomography scans were performed at baseline and at 10 weeks after vaccination. Safety of MVA85A was assessed by clinical, radiological, and inflammatory markers. The immunogenicity of MVA85A was assessed by IFNgamma and IL-2 ELISpot assays and FACS. MEASUREMENTS AND MAIN RESULTS: MVA85A was safe in subjects with LTBI, with comparable adverse events to previous trials of MVA85A. There were no clinically significant changes in inflammatory markers or thoracic computed tomography scans after vaccination. MVA85A induced a strong antigen-specific IFN-gamma and IL-2 response that was durable for 52 weeks. The magnitude of IFN-gamma response was comparable to previous trials of MVA85A in bacillus Calmette-Guérin-vaccinated individuals. Antigen 85A-specific polyfunctional CD4(+) T cells were detectable prior to vaccination with statistically significant increases in cell numbers after vaccination. CONCLUSIONS: MVA85A is safe and highly immunogenic in individuals with LTBI. These results will facilitate further trials in TB-endemic areas. Clinical trial registered with www.clinicaltrials.gov (NCT00456183).


Asunto(s)
Mycobacterium tuberculosis/inmunología , Vacunas contra la Tuberculosis/efectos adversos , Vacunas contra la Tuberculosis/inmunología , Tuberculosis/inmunología , Tuberculosis/terapia , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Prueba de Tuberculina , Tuberculosis/diagnóstico , Vacunas Sintéticas/efectos adversos , Vacunas Sintéticas/inmunología , Adulto Joven
6.
Immunology ; 128(1): 83-91, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19689738

RESUMEN

Maintenance of T-cell responses is an essential feature in protection from many infectious diseases that must be harnessed in vaccination. The relationship between effector T-cell responses and more durable and highly proliferative T-cell memory, particularly in humans, is not well understood. In this study, effector T-cell responses were measured by overnight ex vivo interferon-gamma (IFN-gamma) enzyme-linked immunosorbent spot-forming cell assay (ELISPOT), whereas memory T cells were measured by 10-day culture followed by IFN-gamma ELISPOT (cultured ELISPOT). We observed a significant correlation between IFN-gamma responses to CD4-stimulatory, but not to CD8-stimulatory, recall antigens measured by these assays, suggesting a divergence in regulation. In vaccine trial participants who received a prime-boost vaccination regimen comprising malaria antigens delivered by poxviruses, there was a correlation between ex vivo and cultured responses on day 7, but not 3 months post-vaccination, with the ratio of cultured : ex vivo response increasing over time. To compare responses revealed by cultured ELISPOT in more detail, tetramers comprising viral recall antigens were used to ascribe effector-memory and central-memory T-cell phenotypes through CCR7 and CD62L costaining. For CD8(+) responses the effector phenotype decreased during the initial culture period and memory populations remained high within the resulting 20-fold to 50-fold increased IFN-gamma-secreting or tetramer(+) population. This was less marked for CD4(+) responses, which had higher starting memory phenotype. Depletion of these central-memory T-cell populations generally ablated responses in cultured ELISPOT and reduced ex vivo responses. This study highlights differences between CD4(+) and CD8(+) effector and memory T cells, and the more complex phenotype of CD4(+) T cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Adolescente , Adulto , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática/métodos , Humanos , Memoria Inmunológica/inmunología , Inmunofenotipificación , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Vacunas contra la Malaria/inmunología , Persona de Mediana Edad , Tuberculina/inmunología , Vacunación , Adulto Joven
7.
Immunobiology ; 224(3): 408-418, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30954271

RESUMEN

Hydrophilic lung surfactant proteins have emerged as key immunomodulators which are potent at the recognition and clearance of pulmonary pathogens. Surfactant protein A (SP-A) is a surfactant-associated innate immune molecule, which is known to interact with a variety of pathogens, and display anti-microbial effects. SP-A, being a carbohydrate pattern recognition molecule, has a wide range of innate immune functions against respiratory pathogens, including influenza A virus (IAV). Some pandemic pH1N1 strains resist neutralization by SP-A due to differences in the N-glycosylation of viral hemagglutinin (HA). Here, we provide evidence, for the first time, that a recombinant form of human SP-A (rfhSP-A), composed of α-helical neck and carbohydrate recognition domains, can actually promote the IAV replication, as observed by an upregulation of M1 expression in lung epithelial cell line, A549, when challenged with pH1N1 and H3N2 IAV subtypes. rfhSP-A (10 µg/ml) bound neuraminidase (NA) (∼60 kDa), matrix protein 1 (M1) (∼25 kDa) and M2 (∼17 kDa) in a calcium dependent manner, as revealed by far western blotting, and direct binding ELISA. However, human full length native SP-A downregulated mRNA expression levels of M1 in A549 cells challenged with IAV subtypes. Furthermore, qPCR analysis showed that transcriptional levels of TNF-α, IL-12, IL-6, IFN-α and RANTES were enhanced following rfhSP-A treatment by both IAV subtypes at 6 h post-IAV infection of A549 lung epithelial cells. In the case of full length SP-A treatment, mRNA expression levels of TNF-α and IL-6 were downregulated during the mid-to-late stage of IAV infection of A549 cells. Multiplex cytokine/chemokine array revealed enhanced levels of both IL-6 and TNF-α due to rfhSP-A treatment in the case of both IAV subtypes tested, while no significant effect was seen in the case of IL-12. Enhancement of IAV infection of pH1N1 and H3N2 subtypes by truncated rfhSP-A, concomitant with infection inhibition by full-length SP-A, appears to suggest that a complete SP-A molecule is required for protection against IAV. This is in contrast to a recombinant form of trimeric lectin domains of human SP-D (rfhSP-D) that acts as an entry inhibitor of IAV.


Asunto(s)
Antivirales/metabolismo , Células Epiteliales/fisiología , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H3N2 del Virus de la Influenza A/fisiología , Gripe Humana/inmunología , Pulmón/patología , Proteína A Asociada a Surfactante Pulmonar/metabolismo , Quimiocina CCL5/metabolismo , Citocinas/metabolismo , Células Epiteliales/virología , Glicosilación , Humanos , Mediadores de Inflamación/metabolismo , Unión Proteica , Dominios Proteicos/genética , Proteína A Asociada a Surfactante Pulmonar/genética , Proteínas Recombinantes/genética , Virulencia , Replicación Viral
8.
Immunobiology ; 224(5): 625-631, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31519376

RESUMEN

The complement system is an important humoral immune surveillance mechanism against tumours. However, many malignant tumours are resistant to complement mediated lysis. Here, we report secretion of complement factor H related protein 5 (FHR5) by primary tumour cells derived from Glioblastoma multiforme (GBM) patients. We investigated whether the secreted FHR5 exhibited functional activity similar to factor H, including inhibition of complement mediated lysis, acting as a co-factor for factor I mediated cleavage of C3b, and decay acceleration of C3 convertase. Immunoblotting analysis of primary GBM cells (B30, B31 and B33) supernatant showed the active secretion of FHR5, but not of Factor H. ELISA revealed that the secretion of soluble GBM-FHR5 by cultured GBM cells increased in a time-dependent manner. Primary GBM-FHR5 inhibited complement mediated lysis, possessed co-factor activity for factor I mediated cleavage and displayed decay acceleration of C3 convertase. In summary, we detected the secretion of FHR5 by primary GBM cells B30, B31 and B33. The results demonstrated that GBM-FHR5 shares biological function with FH as a mechanism primary GBM cells potentially use to resist complement mediated lysis.


Asunto(s)
Proteínas del Sistema Complemento/biosíntesis , Glioblastoma/metabolismo , Biomarcadores , Activación de Complemento , Convertasas de Complemento C3-C5/metabolismo , Complemento C3b/inmunología , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Glioblastoma/inmunología , Hemólisis/inmunología , Humanos , Proteolisis
9.
Front Microbiol ; 9: 1903, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30258409

RESUMEN

Mycobacterium tuberculosis, the causative agent of tuberculosis, is one of the leading causes of human deaths due to a single infectious agent. M. tuberculosis infection of the host initiates a local inflammatory response, resulting in the production of a range of inflammatory factors at the site of infection. These inflammatory factors may come in direct contact with M. tuberculosis and immune cells to activate different signaling pathways. One such factor produced in excess during inflammation is a phospholipid compound, Platelet Activating Factor C-16 (PAF C-16). In this study, PAF C-16 was shown to have a direct inhibitory effect on the growth of Mycobacterium bovis BCG (M. bovis BCG) and Mycobacterium smegmatis (M. smegmatis) in a dose- and time-dependent manner. Use of a range of PAF C-16 structural analogs, including the precursor form Lyso-PAF, revealed that small modifications in the structure of PAF C-16 did not alter its mycobacterial growth inhibitory properties. Subsequent experiments suggested that the attachment of aliphatic carbon tail via ether bond to the glycerol backbone of PAF C-16 was likely to play a vital role in its growth inhibition ability against mycobacteria. Fluorescence microscopy and flow cytometry using Propidium iodide (PI) indicated that PAF C-16 treatment had a damaging effect on the cell membrane of M. bovis BCG and M. smegmatis. Furthermore, the growth inhibitory effect of PAF C-16 was partially mitigated by treatment with membrane-stabilizing agents, α-tocopherol and Tween-80, which further suggests that the growth inhibitory effect of PAF C-16 was mediated through bacterial cell membrane damage.

10.
Front Immunol ; 9: 1586, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30105014

RESUMEN

Surfactant protein D (SP-D) is expressed in the mucosal secretion of the lung and contributes to the innate host defense against a variety of pathogens, including influenza A virus (IAV). SP-D can inhibit hemagglutination and infectivity of IAV, in addition to reducing neuraminidase (NA) activity via its carbohydrate recognition domain (CRD) binding to carbohydrate patterns (N-linked mannosylated) on NA and hemagglutinin (HA) of IAV. Here, we demonstrate that a recombinant fragment of human SP-D (rfhSP-D), containing homotrimeric neck and CRD regions, acts as an entry inhibitor of IAV and downregulates M1 expression considerably in A549 cells challenged with IAV of H1N1 and H3N2 subtypes at 2 h treatment. In addition, rfhSP-D downregulated mRNA levels of TNF-α, IFN-α, IFN-ß, IL-6, and RANTES, particularly during the initial stage of IAV infection of A549 cell line. rfhSP-D also interfered with IAV infection of Madin Darby canine kidney (MDCK) cells through HA binding. Furthermore, rfhSP-D was found to reduce luciferase reporter activity in MDCK cells transduced with H1+N1 pseudotyped lentiviral particles, where 50% of reduction was observed with 10 µg/ml rfhSP-D, suggestive of a critical role of rfhSP-D as an entry inhibitor against IAV infectivity. Multiplex cytokine array revealed that rfhSP-D treatment of IAV challenged A549 cells led to a dramatic suppression of key pro-inflammatory cytokines and chemokines. In the case of pH1N1, TNF-α, IFN-α, IL-10, IL-12 (p40), VEGF, GM-CSF, and eotaxin were considerably suppressed by rfhSP-D treatment at 24 h. However, these suppressive effects on IL-10, VEGF, eotaxin and IL-12 (p40) were not so evident in the case of H3N2 subtype, with the exception of TNF-α, IFN-α, and GM-CSF. These data seem to suggest that the extent of immunomodulatory effect of SP-D on host cells can vary considerably in a IAV subtype-specific manner. Thus, rfhSP-D treatment can downregulate pro-inflammatory milieu encouraged by IAV that otherwise causes aberrant inflammatory cell recruitment leading to cell death and lung damage.


Asunto(s)
Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Proteína D Asociada a Surfactante Pulmonar/inmunología , Células A549 , Animales , Aves , Citocinas/inmunología , Perros , Hemaglutininas/inmunología , Humanos , Células de Riñón Canino Madin Darby , Péptidos/genética , Péptidos/inmunología , Proteína D Asociada a Surfactante Pulmonar/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología
11.
Front Immunol ; 9: 533, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29867915

RESUMEN

Mycobacterium tuberculosis can proficiently enter macrophages and diminish complement activation on its cell surface. Within macrophages, the mycobacterium can suppress macrophage apoptosis and survive within the intracellular environment. Previously, we have shown that complement regulatory proteins such as factor H may interfere with pathogen-macrophage interactions during tuberculosis infection. In this study, we show that Mycobacterium bovis BCG binds properdin, an upregulator of the complement alternative pathway. TSR4+5, a recombinant form of thrombospondin repeats 4 and 5 of human properdin expressed in tandem, which is an inhibitor of the alternative pathway, was also able to bind to M. bovis BCG. Properdin and TSR4+5 were found to inhibit uptake of M. bovis BCG by THP-1 macrophage cells in a dose-dependent manner. Quantitative real-time PCR revealed elevated pro-inflammatory responses (TNF-α, IL-1ß, and IL-6) in the presence of properdin or TSR4+5, which gradually decreased over 6 h. Correspondingly, anti-inflammatory responses (IL-10 and TGF-ß) showed suppressed levels of expression in the presence of properdin, which gradually increased over 6 h. Multiplex cytokine array analysis also revealed that properdin and TSR4+5 significantly enhanced the pro-inflammatory response (TNF-α, IL-1ß, and IL-1α) at 24 h, which declined at 48 h, whereas the anti-inflammatory response (IL-10) was suppressed. Our results suggest that properdin may interfere with mycobacterial entry into macrophages via TSR4 and TSR5, particularly during the initial stages of infection, thus affecting the extracellular survival of the pathogen. This study offers novel insights into the non-complement related functions of properdin during host-pathogen interactions in tuberculosis.


Asunto(s)
Macrófagos/fisiología , Mycobacterium bovis/fisiología , Properdina/fisiología , Trombospondinas/fisiología , Citocinas/genética , Humanos , Células THP-1
12.
Clin Vaccine Immunol ; 24(3)2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28077441

RESUMEN

Macaques play a central role in the development of human tuberculosis (TB) vaccines. Immune and challenge responses differ across macaque and human subpopulations. We used novel immunostimulation/immunodynamic modeling methods in a proof-of-concept study to determine which macaque subpopulations best predicted immune responses in different human subpopulations. Data on gamma interferon (IFN-γ)-secreting CD4+ T cells over time after recent Mycobacterium bovis BCG vaccination were available for 55 humans and 81 macaques. Human population covariates were baseline BCG vaccination status, time since BCG vaccination, gender, and the monocyte/lymphocyte cell count ratio. The macaque population covariate was the colony of origin. A two-compartment mathematical model describing the dynamics of the IFN-γ T cell response after BCG vaccination was calibrated to these data using nonlinear mixed-effects methods. The model was calibrated to macaque and human data separately. The association between subpopulations and the BCG immune response in each species was assessed. The macaque subpopulations that best predicted immune responses in different human subpopulations were identified using Bayesian information criteria. We found that the macaque colony and the human baseline BCG status were significantly (P < 0.05) associated with the BCG-induced immune response. For humans who were BCG naïve at baseline, Indonesian cynomolgus macaques and Indian rhesus macaques best predicted the immune response. For humans who had already been BCG vaccinated at baseline, Mauritian cynomolgus macaques best predicted the immune response. This work suggests that the immune responses of different human populations may be best modeled by different macaque colonies, and it demonstrates the potential utility of immunostimulation/immunodynamic modeling to accelerate TB vaccine development.


Asunto(s)
Vacuna BCG/administración & dosificación , Vacuna BCG/inmunología , Linfocitos T CD4-Positivos/inmunología , Descubrimiento de Drogas/métodos , Voluntarios Sanos , Interferón gamma/metabolismo , Mycobacterium bovis/inmunología , Adolescente , Adulto , Animales , Niño , Preescolar , Femenino , Humanos , Lactante , Macaca , Masculino , Persona de Mediana Edad , Modelos Teóricos , Adulto Joven
13.
Clin Cancer Res ; 11(18): 6686-94, 2005 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16166448

RESUMEN

PURPOSE: Alpha-fetoprotein (AFP) is a tumor-associated antigen in hepatocellular carcinoma and is a target for the development of cancer vaccine. Four immunodominant AFP-derived HLA-A*0201-restricted peptides have been identified and the administration of these peptides with an adjuvant has stimulated AFP-specific CTL responses in hepatocellular carcinoma patients. However, no AFP-derived CD4 T-cell epitope has yet been reported and the status of AFP-specific CD4(+) T-cell responses in hepatocellular carcinoma patients is not fully understood. The aim of this study was to analyze naturally occurring CD4(+) T-cell responses to AFP. EXPERIMENTAL DESIGN: We analyzed the ability of CD4(+) T cells to recognize an HLA-DR-restricted AFP-derived epitope in 41 hepatocellular carcinoma patients and 24 non-hepatocellular carcinoma control patients using intracellular cytokine assays for IFN-gamma. RESULTS: Here, for the first time, we report the identification of an AFP-derived CD4(+) T-cell epitope that is recognized by circulating lymphocytes from hepatocellular carcinoma patients in association with HLA-DR. The absence of detectable responses in healthy donors and patients with chronic liver disease suggests that AFP-specific CD4(+) T cells in the responder patients had been previously expanded in vivo in response to the tumor. The anti-AFP CD4(+) T-cell response was only detected in hepatocellular carcinoma patients with normal or mildly elevated serum AFP levels who were in the early stage of disease. CONCLUSION: Our data will be instrumental in the development of cancer vaccine using AFP-derived immunogens.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Carcinoma Hepatocelular/sangre , Epítopos/inmunología , Neoplasias Hepáticas/sangre , alfa-Fetoproteínas/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Secuencia de Aminoácidos , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/inmunología , Células Cultivadas , Epítopos/genética , Femenino , Citometría de Flujo , Antígenos HLA-DR/inmunología , Humanos , Interferón gamma/biosíntesis , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/inmunología , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , alfa-Fetoproteínas/genética
14.
Front Immunol ; 7: 599, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28066412

RESUMEN

Complement protein C1q is the first recognition subcomponent of the complement classical pathway that plays a vital role in the clearance of immune complexes, pathogens, and apoptotic cells. C1q also has a homeostatic role involving immune and non-immune cells; these functions not necessarily involve complement activation. Recently, C1q has been shown to be expressed locally in the microenvironment of a range of human malignant tumors, where it can promote cancer cell adhesion, migration, and proliferation, without involving complement activation. C1q has been shown to be present in the ascitic fluid formed during ovarian cancers. In this study, we have examined the effects of human C1q and its globular domain on an ovarian cancer cell line, SKOV3. We show that C1q and the recombinant globular head modules induce apoptosis in SKOV3 cells in a time-dependent manner. C1q expression was not detectable in the SKOV3 cells. Exogenous treatment with C1q and globular head modules at the concentration of 10 µg/ml induced apoptosis in approximately 55% cells, as revealed by immunofluorescence microscopy and FACS. The qPCR and caspase analysis suggested that C1q and globular head modules activated tumor necrosis factor (TNF)-α and upregulated Fas. The genes of mammalian target of rapamycin (mTOR), RICTOR, and RAPTOR survival pathways, which are often overexpressed in majority of the cancers, were significantly downregulated within few hours of the treatment of SKOV3 cells with C1q and globular head modules. In conclusion, C1q, via its globular domain, induced apoptosis in an ovarian cancer cell line SKOV3 via TNF-α induced apoptosis pathway involving upregulation of Bax and Fas. This study highlights a potentially protective role of C1q in certain cancers.

15.
Tuberculosis (Edinb) ; 96: 37-43, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26786653

RESUMEN

INTRODUCTION: A more effective tuberculosis (TB) vaccine is needed to eliminate TB disease. Many new vaccine candidates enhance the immunogenicity of the existing vaccine, Bacillus Calmette-Guérin (BCG). Understanding BCG induced immune variation is key to developing a new vaccine. AIMS: We aimed to establish if individual-level covariates were associated with cell-mediated immune response (interferon gamma (IFN-γ)) at vaccine trial enrolment (baseline) in a long-term retrospective analysis (LTR) and after BCG vaccination in a short-term prospective analysis (STP). METHODS: Four covariates were analysed: gender, country, BCG vaccination history and monocyte/lymphocyte cell count ratio. Univariable and multivariable linear regression were conducted on IFN-γ response at baseline for LTR, and area under the curve (AUC), 24 week and peak IFN-γ response for STP. RESULTS: Previous BCG vaccination was strongly associated with higher IFN-γ response at baseline (LTR analysis) (p-values < 0.05). Being male showed a weak association with higher baseline response (p-value = 0.1). BCG revaccination was strongly associated with a larger response increase than primary-vaccination (AUC & peak p-values < 0.01), but did not differ at 24 weeks (STP analysis). All other covariates were non-significant (p-values > 0.1). CONCLUSION: This analysis suggests that previous BCG vaccination and gender are associated with durable IFN-γ responses. Vaccine trials may need to stratify by BCG vaccination history and gender.


Asunto(s)
Vacuna BCG/inmunología , Inmunidad Celular , Interferón gamma/inmunología , Mycobacterium tuberculosis/inmunología , Vacunación , Adolescente , Adulto , Área Bajo la Curva , Vacuna BCG/administración & dosificación , Biomarcadores/sangre , Estudios Transversales , Ensayo de Immunospot Ligado a Enzimas , Femenino , Humanos , Esquemas de Inmunización , Interferón gamma/sangre , Ensayos de Liberación de Interferón gamma , Modelos Lineales , Masculino , Persona de Mediana Edad , Análisis Multivariante , Valor Predictivo de las Pruebas , Estudios Prospectivos , Curva ROC , Estudios Retrospectivos , Factores Sexuales , Factores de Tiempo , Adulto Joven
16.
Front Immunol ; 7: 567, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28018340

RESUMEN

The heterotrimeric globular head (gC1q) domain of human C1q is made up of the C-terminal ends of the three individual chains, ghA, ghB, and ghC. A candidate receptor for the gC1q domain is a multi-functional pattern recognition protein, gC1qR. Since understanding of gC1qR and gC1q interaction could provide an insight into the pleiotropic functions of gC1qR, this study was undertaken to identify the gC1qR-binding site on the gC1q domain, using the recombinant ghA, ghB, and ghC modules and their substitution mutants. Our results show that ghA, ghB, and ghC modules can interact with gC1qR independently, thus reinforcing the notion of modularity within the gC1q domain of human C1q. Mutational analysis revealed that while Arg162 in the ghA module is central to interaction between gC1qR and C1q, a single amino acid substitution (arginine to glutamate) in residue 114 of the ghB module resulted in enhanced binding. Expression of gC1qR and C1q in adherent monocytes with or without pro-inflammatory stimuli was also analyzed by qPCR; it showed an autocrine/paracrine basis of C1q and gC1qR interaction. Microscopic studies revealed that C1q and gC1qR are colocalized on PBMCs. Cell proliferation assays indicated that ghA, ghB, and ghC modules were able to attenuate phytohemagglutinin-stimulated proliferation of PBMCs. Addition of gC1qR had an additive effect on the anti-proliferative effect of globular head modules. In summary, our results identify residues involved in C1q-gC1qR interaction and explain, to a certain level, their involvement on the immune cell surface, which is relevant for C1q-induced functions including inflammation, infection, and immunity.

17.
J Biomed Nanotechnol ; 12(1): 197-216, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27301184

RESUMEN

Nanoparticles are attractive drug delivery vehicles for targeted organ-specific as well as systemic therapy. However, their interaction with the immune system offers an intriguing challenge to the success of nanotherapeutics in vivo. Recently, we showed that pristine and derivatised carbon nanotubes (CNT) can activate complement mainly via the classical pathway leading to enhanced uptake by phagocytic cells, and transcriptional down-regulation of pro-inflammatory cytokines. Here, we report the interaction of complement-activating CC-CNT and RNA-CNT, and non-complement-activating gold-nickel (Au-Ni) nanowires with cell lines representing macrophage, B and T cells. Complement deposition considerably enhanced uptake of CNTs by immune cells known to overexpress complement receptors. Real-Time qPCR and multiplex array analyses showed complement-dependent down-regulation of TNF-α and IL-1ß and up-regulation of IL-12 by CMC- and RNA-CNTs, in addition to revealing IL-10 as a crucial regulator during nanoparticle-immune cell interaction. It appears that complement system can recognize molecular patterns differentially displayed by nanoparticles and thus, modulate subsequent processing of nanoparticles by antigen capturing and antigen presenting cells, which can shape innate and adaptive immune axes.


Asunto(s)
Linfocitos B/inmunología , Citocinas/inmunología , Inmunidad Celular/inmunología , Macrófagos/inmunología , Nanotubos de Carbono/toxicidad , Linfocitos T/inmunología , Linfocitos B/efectos de los fármacos , Línea Celular , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/toxicidad , Humanos , Inmunidad Celular/efectos de los fármacos , Macrófagos/efectos de los fármacos , Ensayo de Materiales , Nanotubos de Carbono/química , Linfocitos T/efectos de los fármacos
18.
Lancet ; 364(9452): 2196-203, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15610806

RESUMEN

BACKGROUND: Childhood tuberculosis often presents non-specifically and is a common differential diagnosis in high prevalence areas. Current diagnostic tools have poor sensitivity and cannot reliably exclude tuberculosis, so overdiagnosis is common. HIV co-infection exacerbates this problem and accounts for an increasing proportion of paediatric tuberculosis worldwide. METHODS: We assessed the usefulness of a T-cell-based rapid blood test for Mycobacterium tuberculosis infection, the enzyme-linked immunospot assay (ELISPOT), in routine clinical practice. We did a prospective blinded study of 293 African children with suspected tuberculosis in kwaZulu-Natal, a region with high HIV prevalence. Children had full clinical assessment, ELISPOT, and a tuberculin skin test. Test results were compared with final clinical and microbiological diagnoses. RESULTS: In children with tuberculosis, sensitivity of ELISPOT was 83% (95% CI 75-89, n=133), significantly higher (p<0.001) than the 63% (54-72) sensitivity of tuberculin skin test (n=116). Sensitivity of tuberculin skin test fell significantly in children younger than 3 years (to 51%), with HIV co-infection (36%), or with malnutrition (44%). Sensitivity of ELISPOT, which was not significantly adversely affected by these factors, was 85%, 73%, and 78%, respectively in these subgroups. In 116 children with both test results available, sensitivity of the two tests combined was 91% (85-95). CONCLUSIONS: Diagnostic sensitivity of ELISPOT is higher than that of the skin test and is less affected by factors frequently associated with childhood tuberculosis in developing countries. Used together with the skin test, ELISPOT provides a clinically useful diagnostic sensitivity in African children with suspected tuberculosis.


Asunto(s)
Antígenos Bacterianos/sangre , Ensayo de Inmunoadsorción Enzimática , Linfocitos T/microbiología , Tuberculosis Pulmonar/diagnóstico , Adolescente , Niño , Preescolar , Femenino , Seropositividad para VIH/complicaciones , Humanos , Lactante , Masculino , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/aislamiento & purificación , Sensibilidad y Especificidad , Sudáfrica , Prueba de Tuberculina , Tuberculosis Pulmonar/complicaciones
19.
Tuberculosis (Edinb) ; 85(1-2): 47-52, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15687027

RESUMEN

There is an urgent need for an improved vaccine against tuberculosis. Heterologous prime-boost immunization regimes induce higher levels of cellular immunity than homologous boosting with the same vaccine. Using BCG as the priming immunization in such a regime allows for the retention of the beneficial protective effects of BCG against disseminated disease in childhood. Recombinant poxviruses are powerful boosting agents, for both CD4+ and CD8+ T cells. Here we review the preclinical data from a BCG prime-recombinant modified vaccinia virus Ankara expressing antigen 85A (MVA85A) boost strategy. MVA85A is now in clinical trials in the UK and Africa and the design of these trials, including the ethical and regulatory issues are discussed.


Asunto(s)
Vacuna BCG/inmunología , Vacunas contra la Tuberculosis/inmunología , Tuberculosis/inmunología , Virus Vaccinia/inmunología , Vacuna BCG/uso terapéutico , Ensayos Clínicos como Asunto , Humanos , Inmunidad Celular , Mycobacterium tuberculosis/inmunología , Linfocitos T/inmunología , Tuberculosis/prevención & control , Vacunas contra la Tuberculosis/uso terapéutico , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/uso terapéutico
20.
AIDS ; 16(17): 2285-93, 2002 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-12441800

RESUMEN

OBJECTIVES: An accurate test for Mycobacterium tuberculosis infection is urgently needed. The tuberculin skin test (TST) lacks sensitivity, particularly in HIV-infected individuals, and has poor specificity because of antigenic cross-reactivity with Bacillus Calmette-Guérin (BCG) vaccination. ESAT-6 and CFP-10 are antigens expressed in Mycobacterium tuberculosis, but not in Mycobacterium bovis BCG and most environmental mycobacteria. We investigated whether T cells specific for these antigens could serve as accurate markers of M. tuberculosis infection in an area of high tuberculosis and HIV prevalence. METHODS: Using the rapid ex-vivo enzyme-linked immunospot (ELISPOT) assay for IFN-gamma, we enumerated T cells specific for ESAT-6, CFP-10 and purified protein derivative (PPD) in blood samples from 50 Zambian tuberculosis patients, 75 healthy Zambian adults, and 40 healthy UK residents. TSTs were performed in 49 healthy Zambian adults. RESULTS: All (100%; n = 11) and 90% (n = 39) of HIV-negative and HIV-positive tuberculosis patients, respectively, had detectable ESAT-6- or CFP-10-specific T cells. The ESAT-6/CFP-10-based ELISPOT assay was positive in 37 out of 54 HIV-negative healthy Zambians, suggesting a 69% prevalence of latent M. tuberculosis infection. Fewer HIV-positive Zambians possessed ESAT-6/CFP-10-specific T cells, but the impact of HIV infection was less on this assay than on the PPD-based ELISPOT or TST. CONCLUSION: The ESAT-6/CFP-10-based ELISPOT assay detects active tuberculosis in HIV-positive individuals with high sensitivity. It is more specific, and possibly more sensitive, than PPD-based methods of detecting latent M. tuberculosis infection, and may potentially improve the targeting of isoniazid preventative therapy to HIV-positive individuals with latent tuberculosis infection.


Asunto(s)
Infecciones por VIH/complicaciones , Mycobacterium tuberculosis/inmunología , Linfocitos T/inmunología , Tuberculosis Pulmonar/diagnóstico , Adolescente , Adulto , Anciano , Antígenos Bacterianos/inmunología , Vacuna BCG , Proteínas Bacterianas/inmunología , Ensayo de Inmunoadsorción Enzimática/métodos , Mapeo Epitopo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mycobacterium tuberculosis/fisiología , Estudios Prospectivos , Sensibilidad y Especificidad , Prueba de Tuberculina , Tuberculosis Pulmonar/complicaciones , Tuberculosis Pulmonar/inmunología , Latencia del Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA