Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
MAbs ; 13(1): 1953220, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34288809

RESUMEN

Pathogens frequently use multivalent binding to sialic acid to infect cells or to modulate immunity through interactions with human sialic acid-binding immunoglobulin-type lectins (Siglecs). Molecules that interfere with these interactions could be of interest as diagnostics, anti-infectives or as immune modulators. This review describes the development of molecular scaffolds based on the crystallizable fragment (Fc) region of immunoglobulin (Ig) G that deliver high-avidity binding to innate immune receptors, including sialic acid-dependent receptors. The ways in which the sialylated Fc may be engineered as immune modulators that mimic the anti-inflammatory properties of intravenous polyclonal Ig or as blockers of sialic-acid-dependent infectivity by viruses are also discussed.


Asunto(s)
Fragmentos Fc de Inmunoglobulinas , Inmunoglobulina G , Ácido N-Acetilneuramínico , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/inmunología , Animales , Glicosilación , Humanos , Fragmentos Fc de Inmunoglobulinas/inmunología , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Inmunoglobulina G/inmunología , Inmunoglobulina G/uso terapéutico , Ácido N-Acetilneuramínico/inmunología , Ácido N-Acetilneuramínico/uso terapéutico
2.
Immunology ; 164(1): 90-105, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33880776

RESUMEN

Intravenous immunoglobulin (IVIG) is an established treatment for numerous autoimmune conditions. Although Fc fragments derived from IVIG have shown efficacy in controlling immune thrombocytopenia in children, the mechanisms of action are unclear and controversial. The aim of this study was to dissect IVIG effector mechanisms using further adapted Fc fragments on demyelination in an ex vivo model of the central nervous system-immune interface. Using organotypic cerebellar slice cultures (OSCs) from transgenic mice, we induced extensive immune-mediated demyelination and oligodendrocyte loss with an antibody specific for myelin oligodendrocyte glycoprotein (MOG) and complement. Protective effects of adapted Fc fragments were assessed by live imaging of green fluorescent protein expression, immunohistochemistry and confocal microscopy. Cysteine- and glycan-adapted Fc fragments protected OSC from demyelination in a dose-dependent manner where equimolar concentrations of either IVIG or control Fc were ineffective. The protective effects of the adapted Fc fragments are partly attributed to interference with complement-mediated oligodendroglia damage. Transcriptome analysis ruled out signatures associated with inflammatory or innate immune responses. Taken together, our findings show that recombinant biomimetics can be made that are at least two hundred-fold more effective than IVIG in controlling demyelination by anti-MOG antibodies.


Asunto(s)
Autoanticuerpos/uso terapéutico , Cerebelo/patología , Enfermedades Desmielinizantes/terapia , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Inmunoglobulina G/uso terapéutico , Oligodendroglía/patología , Proteínas Recombinantes de Fusión/uso terapéutico , Animales , Autoanticuerpos/genética , Cerebelo/efectos de los fármacos , Enfermedades Desmielinizantes/inmunología , Células HEK293 , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoglobulina G/genética , Inmunoglobulinas Intravenosas/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/inmunología , Oligodendroglía/efectos de los fármacos , Técnicas de Cultivo de Órganos , Proteínas Recombinantes de Fusión/genética
3.
J Immunol ; 204(4): 1022-1034, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31907284

RESUMEN

Abs are glycoproteins that carry a conserved N-linked carbohydrate attached to the Fc whose presence and fine structure profoundly impacts on their in vivo immunogenicity, pharmacokinetics, and functional attributes. The host cell line used to produce IgG plays a major role in this glycosylation, as different systems express different glycosylation enzymes and transporters that contribute to the specificity and heterogeneity of the final IgG-Fc glycosylation profile. In this study, we compare two panels of glycan-adapted IgG1-Fc mutants expressed in either the human endothelial kidney 293-F or Chinese hamster ovary-K1 systems. We show that the types of N-linked glycans between matched pairs of Fc mutants vary greatly and in particular, with respect, to sialylation. These cell line effects on glycosylation profoundly influence the ability of the engineered Fcs to interact with either human or pathogen receptors. For example, we describe Fc mutants that potently disrupted influenza B-mediated agglutination of human erythrocytes when expressed in Chinese hamster ovary-K1, but not in human endothelial kidney 293-F cells.


Asunto(s)
Fragmentos Fc de Inmunoglobulinas/metabolismo , Inmunoglobulina G/metabolismo , Virus de la Influenza B/inmunología , Gripe Humana/tratamiento farmacológico , Animales , Especificidad de Anticuerpos , Células CHO , Cricetinae , Cricetulus , Glicosilación , Células HEK293 , Humanos , Fragmentos Fc de Inmunoglobulinas/inmunología , Fragmentos Fc de Inmunoglobulinas/aislamiento & purificación , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Inmunoglobulina G/inmunología , Inmunoglobulina G/aislamiento & purificación , Inmunoglobulina G/uso terapéutico , Gripe Humana/inmunología , Gripe Humana/virología
4.
J Immunol ; 202(5): 1595-1611, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30683699

RESUMEN

In therapeutic applications in which the Fc of IgG is critically important, the receptor binding and functional properties of the Fc are lost after deglycosylation or removal of the unique Asn297 N-X-(T/S) sequon. A population of Fcs bearing sialylated glycans has been identified as contributing to this functionality, and high levels of sialylation also lead to longer serum retention times advantageous for therapy. The efficacy of sialylated Fc has generated an incentive to modify the unique N-linked glycosylation site at Asn297, either through chemical and enzymatic methods or by mutagenesis of the Fc, that disrupts the protein-Asn297 carbohydrate interface. In this study, we took an alternative approach by inserting or deleting N-linked attachment sites into the body of the Fc to generate a portfolio of mutants with tailored effector functions. For example, we describe mutants with enhanced binding to low-affinity inhibitory human Fcγ and glycan receptors that may be usefully incorporated into existing Ab engineering approaches to treat or vaccinate against disease. The IgG1 Fc fragments containing complex sialylated glycans attached to the N-terminal Asn221 sequon bound influenza virus hemagglutinin and disrupted influenza A-mediated agglutination of human erythrocytes.


Asunto(s)
Hemaglutinación/genética , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoglobulina G/genética , Orthomyxoviridae/genética , Polisacáridos/genética , Receptores de IgG/genética , Glicosilación , Hemaglutinación/inmunología , Humanos , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina G/inmunología , Mutación , Orthomyxoviridae/inmunología , Polisacáridos/inmunología , Receptores de IgG/inmunología
5.
J Biol Chem ; 292(31): 12994-13007, 2017 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-28620050

RESUMEN

Multimeric fragment crystallizable (Fc) regions and Fc-fusion proteins are actively being explored as biomimetic replacements for IVIG therapy, which is deployed to manage many diseases and conditions but is expensive and not always efficient. The Fc region of human IgG1 (IgG1-Fc) can be engineered into multimeric structures (hexa-Fcs) that bind their cognate receptors with high avidity. The critical influence of the unique N-linked glycan attached at Asn-297 on the structure and function of IgG1-Fc is well documented; however, whether the N-linked glycan has a similarly critical role in multimeric, avidly binding Fcs, is unknown. Hexa-Fc contains two N-linked sites at Asn-77 (equivalent to Asn-297 in the Fc of IgG1) and Asn-236 (equivalent to Asn-563 in the tail piece of IgM). We report here that glycosylation at Asn-297 is critical for interactions with Fc receptors and complement and that glycosylation at Asn-563 is essential for controlling multimerization. We also found that introduction of an additional fully occupied N-linked glycosylation site at the N terminus at position 1 (equivalent to Asp-221 in the Fc of IgG1) dramatically enhances overall sialic acid content of the Fc multimers. Furthermore, replacement of Cys-575 in the IgM tail piece of multimers resulted in monomers with enhanced sialic acid content and differential receptor-binding profiles. Thus insertion of additional N-linked glycans into either the hinge or tail piece of monomers or multimers leads to molecules with enhanced sialylation that may be suitable for managing inflammation or blocking pathogen invasion.


Asunto(s)
Diseño de Fármacos , Fragmentos Fc de Inmunoglobulinas/metabolismo , Inmunoglobulina G/metabolismo , Modelos Moleculares , Ingeniería de Proteínas , Procesamiento Proteico-Postraduccional , Receptores de Superficie Celular/metabolismo , Sustitución de Aminoácidos , Animales , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/metabolismo , Asparagina/metabolismo , Células CHO , Cricetulus , Cistina/metabolismo , Glicosilación , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoglobulina G/química , Inmunoglobulina G/genética , Estructura Molecular , Peso Molecular , Mutagénesis Sitio-Dirigida , Mutación , Dominios y Motivos de Interacción de Proteínas , Receptores de Superficie Celular/química , Receptores de Superficie Celular/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
6.
Sci Rep ; 7: 42989, 2017 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-28230186

RESUMEN

IgM is the first antibody to be produced in immune responses and plays an important role in the neutralization of bacteria and viruses. Human IgM is heavily glycosylated, featuring five N-linked glycan sites on the µ chain and one on the J-chain. Glycosylation of IgG is known to modulate the effector functions of Fcγ receptors. In contrast, little is known about the effect of glycosylation on IgM binding to the human Fcµ receptor (hFCMR). In this study, we identify the Cµ4 domain of IgM as the target of hFCMR, and show that binding and internalization of IgM by hFCMR is glycan-independent. We generated a homology-based structure for hFCMR and used molecular dynamic simulations to show how this interaction with IgM may occur. Finally, we reveal an inhibitory function for IgM in the proliferation of T cells.


Asunto(s)
Inmunoglobulina M/metabolismo , Polisacáridos/química , Receptores Fc/metabolismo , Sitios de Unión , Proliferación Celular/efectos de los fármacos , Endocitosis , Glicosilación , Humanos , Inmunoglobulina M/química , Inmunoglobulina M/farmacología , Simulación de Dinámica Molecular , Fitohemaglutininas/farmacología , Unión Proteica , Estructura Terciaria de Proteína , Receptores Fc/química , Linfocitos T/citología , Linfocitos T/metabolismo
7.
J Biol Chem ; 291(27): 14285-14299, 2016 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-27226583

RESUMEN

Diversity at pathogen genetic loci can be driven by host adaptive immune selection pressure and may reveal proteins important for parasite biology. Population-based genome sequencing of Plasmodium falciparum, the parasite responsible for the most severe form of malaria, has highlighted two related polymorphic genes called dblmsp and dblmsp2, which encode Duffy binding-like (DBL) domain-containing proteins located on the merozoite surface but whose function remains unknown. Using recombinant proteins and transgenic parasites, we show that DBLMSP and DBLMSP2 directly and avidly bind human IgM via their DBL domains. We used whole genome sequence data from over 400 African and Asian P. falciparum isolates to show that dblmsp and dblmsp2 exhibit extreme protein polymorphism in their DBL domain, with multiple variants of two major allelic classes present in every population tested. Despite this variability, the IgM binding function was retained across diverse sequence representatives. Although this interaction did not seem to have an effect on the ability of the parasite to invade red blood cells, binding of DBLMSP and DBLMSP2 to IgM inhibited the overall immunoreactivity of these proteins to IgG from patients who had been exposed to the parasite. This suggests that IgM binding might mask these proteins from the host humoral immune system.


Asunto(s)
Antígenos de Protozoos/metabolismo , Inmunoglobulina M/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Animales , Humanos , Unión Proteica
8.
Sci Rep ; 6: 20509, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-26842628

RESUMEN

Several splice variants of IgE exist in human plasma, including a variant called IgE-tailpiece (IgE-tp) that differs from classical IgE by the replacement of two carboxy-terminal amino acids with eight novel residues that include an ultimate cysteine. To date, the role of the secreted IgE-tp isoform in human immunity is unknown. We show that levels of IgE-tp are raised in helminth-infected donors, and that both the classical form of IgE (IgE-c) and IgE-tp interact with polymers of the serine protease inhibitor alpha-1-antitrypsin (A1AT). The association of IgE-tp with A1AT polymers in plasma protects the antibody from serine protease-mediated degradation, without affecting the functional interaction of IgE-tp with important receptors, including FcεR1. That polymers of A1AT protect IgE from degradation by helminth proteases may explain why these common and normally non-disease causing polymorphic variants of A1AT have been retained by natural selection. The observation that IgE can be complexed with polymeric forms of A1AT may therefore have important consequences for our understanding of the pathophysiology of pulmonary diseases that arise either as a consequence of A1AT-deficiency or through IgE-mediated type 1 hypersensitivity responses.


Asunto(s)
Inmunoglobulina E/metabolismo , Receptores de IgE/metabolismo , alfa 1-Antitripsina/metabolismo , Helmintiasis/inmunología , Humanos , Inmunoglobulina E/química , Isoformas de Proteínas/metabolismo , Proteolisis
9.
J Control Release ; 223: 42-52, 2016 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-26718855

RESUMEN

Monoclonal IgG antibodies (Abs) are used extensively in the clinic to treat cancer and autoimmune diseases. In addition, therapeutic proteins are genetically fused to the constant Fc part of IgG. In both cases, the Fc secures a long serum half-life and favourable pharmacokinetics due to its pH-dependent interaction with the neonatal Fc receptor (FcRn). FcRn also mediates transport of intact IgG across polarized epithelial barriers, a pathway that is attractive for delivery of Fc-containing therapeutics. So far, no study has thoroughly compared side-by-side how IgG and different Fc-fusion formats are transported across human polarizing epithelial cells. Here, we used an in vitro cellular transport assay based on the human polarizing epithelial cell line (T84) in which both IgG1 and Fc-fusions were transported in an FcRn-dependent manner. Furthermore, we found that the efficacy of transport was dependent on the format. We demonstrate that transepithelial delivery could be enhanced by Fc-engineering for improved FcRn binding as well as by Fc-polymerization. In both cases, transport was driven by pH-dependent binding kinetics and the pH at the luminal side. Hence, efficient transcellular delivery of IgG-based drugs across human epithelial cells requires optimal pH-dependent FcRn binding that can be manipulated by avidity and Fc-engineering, factors that should inspire the design of future therapeutics targeted for transmucosal delivery.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Fragmentos Fc de Inmunoglobulinas/metabolismo , Inmunoglobulina G/metabolismo , Receptores Fc/metabolismo , Anticuerpos Monoclonales/metabolismo , Línea Celular Tumoral , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoglobulina G/genética , Polimerizacion , Ingeniería de Proteínas , Receptores Fc/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
10.
Trends Parasitol ; 32(2): 108-119, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26597020

RESUMEN

Immunoglobulin M (IgM) is an ancient antibody class that is found in all vertebrates, with the exception of coelacanths, and is indispensable in both innate and adaptive immunity. The equally ancient human malaria parasite, Plasmodium falciparum, formed an intimate relationship with IgM with which it co-evolved. In this article, we discuss the association between IgM and human malaria parasites, building on several recent publications that implicate IgM as a crucial molecule that determines both host and parasite survival. Consequently, a better understanding of this association may lead to the development of improved intervention strategies.


Asunto(s)
Interacciones Huésped-Parásitos , Evasión Inmune/inmunología , Inmunoglobulina M/inmunología , Malaria Falciparum/inmunología , Animales , Humanos , Inflamación/inmunología , Plasmodium falciparum/inmunología
11.
Sci Rep ; 5: 14081, 2015 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-26373337

RESUMEN

The assessment of naturally-acquired and vaccine-induced immunity to blood-stage Plasmodium falciparum malaria is of long-standing interest. However, the field has suffered from a paucity of in vitro assays that reproducibly measure the anti-parasitic activity induced by antibodies in conjunction with immune cells. Here we optimize the antibody-dependent respiratory burst (ADRB) assay, which assesses the ability of antibodies to activate the release of reactive oxygen species from human neutrophils in response to P. falciparum blood-stage parasites. We focus particularly on assay parameters affecting serum preparation and concentration, and importantly assess reproducibility. Our standardized protocol involves testing each serum sample in singlicate with three independent neutrophil donors, and indexing responses against a standard positive control of pooled hyper-immune Kenyan sera. The protocol can be used to quickly screen large cohorts of samples from individuals enrolled in immuno-epidemiological studies or clinical vaccine trials, and requires only 6 µL of serum per sample. Using a cohort of 86 samples, we show that malaria-exposed individuals induce higher ADRB activity than malaria-naïve individuals. The development of the ADRB assay complements the use of cell-independent assays in blood-stage malaria, such as the assay of growth inhibitory activity, and provides an important standardized cell-based assay in the field.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Neutrófilos/inmunología , Plasmodium falciparum/inmunología , Estallido Respiratorio/inmunología , Adulto , Humanos , Técnicas In Vitro , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Neutrófilos/metabolismo , Receptores Fc/metabolismo , Reproducibilidad de los Resultados
12.
Sci Rep ; 5: 9526, 2015 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-25912958

RESUMEN

The remarkable clinical success of Fc-fusion proteins has driven intense investigation for even more potent replacements. Using quality-by-design (QbD) approaches, we generated hexameric-Fc (hexa-Fc), a ~20 nm oligomeric Fc-based scaffold that we here show binds low-affinity inhibitory receptors (FcRL5, FcγRIIb, and DC-SIGN) with high avidity and specificity, whilst eliminating significant clinical limitations of monomeric Fc-fusions for vaccine and/or cancer therapies, in particular their poor ability to activate complement. Mass spectroscopy of hexa-Fc reveals high-mannose, low-sialic acid content, suggesting that interactions with these receptors are influenced by the mannose-containing Fc. Molecular dynamics (MD) simulations provides insight into the mechanisms of hexa-Fc interaction with these receptors and reveals an unexpected orientation of high-mannose glycans on the human Fc that provides greater accessibility to potential binding partners. Finally, we show that this biosynthetic nanoparticle can be engineered to enhance interactions with the human neonatal Fc receptor (FcRn) without loss of the oligomeric structure, a crucial modification for these molecules in therapy and/or vaccine strategies where a long plasma half-life is critical.


Asunto(s)
Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulinas Intravenosas/inmunología , Vacunas/inmunología , Biomimética/métodos , Proteínas Portadoras/inmunología , Células Cultivadas , Semivida , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Leucocitos Mononucleares/inmunología , Unión Proteica/inmunología , Receptores Fc/inmunología
13.
Sci Rep ; 4: 3618, 2014 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-24402577

RESUMEN

The major antimalarial drug quinine perturbs uptake of the essential amino acid tryptophan, and patients with low plasma tryptophan are predisposed to adverse quinine reactions; symptoms of which are similar to indications of tryptophan depletion. As tryptophan is a precursor of the neurotransmitter serotonin (5-HT), here we test the hypothesis that quinine disrupts serotonin function. Quinine inhibited serotonin-induced proliferation of yeast as well as human (SHSY5Y) cells. One possible cause of this effect is through inhibition of 5-HT receptor activation by quinine, as we observed here. Furthermore, cells exhibited marked decreases in serotonin production during incubation with quinine. By assaying activity and kinetics of the rate-limiting enzyme for serotonin biosynthesis, tryptophan hydroxylase (TPH2), we showed that quinine competitively inhibits TPH2 in the presence of the substrate tryptophan. The study shows that quinine disrupts both serotonin biosynthesis and function, giving important new insight to the action of quinine on mammalian cells.


Asunto(s)
Antimaláricos/farmacología , Quinina/farmacología , Serotonina/biosíntesis , Línea Celular Tumoral , Humanos , Neuroblastoma/patología , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/crecimiento & desarrollo , Serotonina/fisiología , Triptófano Hidroxilasa/antagonistas & inhibidores
14.
J Leukoc Biol ; 95(2): 369-82, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24163420

RESUMEN

New tools are required to expedite the development of an effective vaccine against the blood-stage infection with the human malaria parasite Plasmodium falciparum. This work describes the assessment of the ADRB assay in a mouse model, characterizing the functional interaction between antimalarial serum antibodies and FcRs upon neutrophils. We describe a reproducible, antigen-specific assay, dependent on functional FcR signaling, and show that ADRB activity is induced equally by IgG1 and IgG2a isotypes and is modulated by blocking FcR function. However, following immunization of mice with the blood-stage vaccine candidate antigen MSP142, no measurable ADRB activity was induced against PEMS and neither was vaccine efficacy modulated against Plasmodium yoelii blood-stage challenge in γ(-/-) mice compared with WT mice. In contrast, following a primary, nonlethal P. yoelii parasite challenge, serum from vaccinated mice and nonimmunized controls showed anti-PEMS ADRB activity. Upon secondary challenge, nonimmunized γ(-/-) mice showed a reduced ability to control blood-stage parasitemia compared with immunized γ(-/-) mice; however, WT mice, depleted of their neutrophils, did not lose their ability to control infection. Thus, whereas neutrophil-induced ADRB against PEMS does not appear to play a role in protection against P. yoelii rodent malaria, induction of ADRB activity after challenge suggests that antigen targets of anti-PEMS ADRB activity remain to be established, as well as further supporting the observation that ADRB activity to P. falciparum arises following repeated natural exposure.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Inmunoensayo/métodos , Malaria/inmunología , Malaria/parasitología , Neutrófilos/inmunología , Plasmodium yoelii/inmunología , Estallido Respiratorio/inmunología , Animales , Antígenos de Protozoos/inmunología , Femenino , Humanos , Inmunoglobulina G/metabolismo , Ratones , Parásitos/inmunología , Receptores Inmunológicos/metabolismo , Transducción de Señal/inmunología , Resultado del Tratamiento
16.
Antimicrob Agents Chemother ; 57(8): 3889-96, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23733464

RESUMEN

Chloroquine (CQ) has been a mainstay of antimalarial drug treatment for several decades. Additional therapeutic actions of CQ have been described, including some reports of fungal inhibition. Here we investigated the action of CQ in fungi, including the yeast model Saccharomyces cerevisiae. A genomewide yeast deletion strain collection was screened against CQ, revealing that bck1Δ and slt2Δ mutants of the cell wall integrity pathway are CQ hypersensitive. This phenotype was rescued with sorbitol, consistent with cell wall involvement. The cell wall-targeting agent caffeine caused hypersensitivity to CQ, as did cell wall perturbation by sonication. The phenotypes were not caused by CQ-induced changes to cell wall components. Instead, CQ accumulated to higher levels in cells with perturbed cell walls: CQ uptake was 2- to 3-fold greater in bck1Δ and slt2Δ mutants than in wild-type yeast. CQ toxicity was synergistic with that of the major cell wall-targeting antifungal drug, caspofungin. The MIC of caspofungin against the yeast pathogen Candida albicans was decreased 2-fold by 250 µM CQ and up to 8-fold at higher CQ concentrations. Similar effects were seen in Candida glabrata and Aspergillus fumigatus. The results show that the cell wall is critical for CQ resistance in fungi and suggest that combination treatments with cell wall-targeting drugs could have potential for antifungal treatment.


Asunto(s)
Antimaláricos/farmacología , Pared Celular/efectos de los fármacos , Cloroquina/farmacología , Farmacorresistencia Fúngica , Saccharomyces cerevisiae/efectos de los fármacos , Aspergillus fumigatus/efectos de los fármacos , Transporte Biológico , Candida albicans/efectos de los fármacos , Candida glabrata/efectos de los fármacos , Caspofungina , Sinergismo Farmacológico , Equinocandinas/farmacología , Lipopéptidos , Pruebas de Sensibilidad Microbiana , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Sorbitol/farmacología
17.
Sci Rep ; 3: 1706, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23609325

RESUMEN

Rodent malaria species Plasmodium yoelii and P. chabaudi have been widely used to validate vaccine approaches targeting blood-stage merozoite antigens. However, increasing data suggest the P. berghei rodent malaria may be able to circumvent vaccine-induced anti-merozoite responses. Here we confirm a failure to protect against P. berghei, despite successful antibody induction against leading merozoite antigens using protein-in-adjuvant or viral vectored vaccine delivery. No subunit vaccine approach showed efficacy in mice following immunization and challenge with the wild-type P. berghei strains ANKA or NK65, or against a chimeric parasite line encoding a merozoite antigen from P. falciparum. Protection was not improved in knockout mice lacking the inhibitory Fc receptor CD32b, nor against a Δsmac P. berghei parasite line with a non-sequestering phenotype. An improved understanding of the mechanisms responsible for protection, or failure of protection, against P. berghei merozoites could guide the development of an efficacious vaccine against P. falciparum.


Asunto(s)
Formación de Anticuerpos/inmunología , Antimaláricos/inmunología , Vacunas contra la Malaria/inmunología , Malaria/inmunología , Merozoítos/inmunología , Plasmodium berghei/inmunología , Animales , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/inmunología , Línea Celular , Femenino , Células HEK293 , Humanos , Inmunización/métodos , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Conejos , Receptores de IgG/inmunología , Roedores/inmunología
18.
Exp Parasitol ; 132(3): 367-72, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22960347

RESUMEN

Acanthamoeba granulomatous encephalitis (AGE), caused by Acanthamoeba castellanii, is a fatal infection of immunocompromised individuals. The pathogenesis of blood-brain barrier (BBB) breach remains unknown. Using a novel in vitro BBB infection model under flow conditions, demonstrates that increases in flow rates lead to decreased binding of A. castellanii to host cells. This is a distinct departure from previous findings under static conditions. However, similarly to static conditions binding of A. castellanii to host cells is host mannose dependent. Disruption of the host cell monolayer was independent of amoeba binding, but dependent on secreted serine proteases. For the first time we report the binding dynamics of A. castellanii under physiological conditions, showing that BBB disruption is not directly linked to binding, instead it is reliant on secreted proteases. Our results offer a platform on which therapies designed at modulating physiological parameters can improve the outcome of infection with A. castellanii.


Asunto(s)
Acanthamoeba castellanii/fisiología , Amebiasis/parasitología , Barrera Hematoencefálica/parasitología , Encefalitis/parasitología , Serina Proteasas/metabolismo , Encéfalo/citología , Encéfalo/parasitología , Células Cultivadas , Medios de Cultivo Condicionados , Células Endoteliales/parasitología , Endotelio Vascular/citología , Endotelio Vascular/parasitología , Humanos , Hidrodinámica , Lectina de Unión a Manosa/metabolismo , Microvasos/metabolismo , Microvasos/parasitología
19.
PLoS One ; 7(9): e44943, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22984589

RESUMEN

Viral vectored vaccines have been shown to induce both T cell and antibody responses in animals and humans. However, the induction of even higher level T cell responses may be crucial in achieving vaccine efficacy against difficult disease targets, especially in humans. Here we investigate the oligomerization domain of the α-chain of C4b-binding protein (C4 bp) as a candidate T cell "molecular adjuvant" when fused to malaria antigens expressed by human adenovirus serotype 5 (AdHu5) vectored vaccines in BALB/c mice. We demonstrate that i) C-terminal fusion of an oligomerization domain can enhance the quantity of antigen-specific CD4(+) and CD8(+) T cell responses induced in mice after only a single immunization of recombinant AdHu5, and that the T cells maintain similar functional cytokine profiles; ii) an adjuvant effect is observed for AdHu5 vectors expressing either the 42 kDa C-terminal domain of Plasmodium yoelii merozoite surface protein 1 (PyMSP1(42)) or the 83 kDa ectodomain of P. falciparum strain 3D7 apical membrane antigen 1 (PfAMA1), but not a candidate 128kDa P. falciparum MSP1 biallelic fusion antigen; iii) following two homologous immunizations of AdHu5 vaccines, antigen-specific T cell responses are further enhanced, however, in both BALB/c mice and New Zealand White rabbits no enhancement of functional antibody responses is observed; and iv) that the T cell adjuvant activity of C4 bp is not dependent on a functional Fc-receptor γ-chain in the host, but is associated with the oligomerization of small (<80 kDa) antigens expressed by recombinant AdHu5. The oligomerization domain of C4 bp can thus adjuvant T cell responses induced by AdHu5 vectors against selected antigens and its clinical utility as well as mechanism of action warrant further investigation.


Asunto(s)
Adenoviridae/genética , Proteína de Unión al Complemento C4b/metabolismo , Malaria/prevención & control , Linfocitos T/citología , Adyuvantes Inmunológicos/genética , Animales , Antígenos de Protozoos/genética , Femenino , Vectores Genéticos , Vacunas contra la Malaria/genética , Proteína 1 de Superficie de Merozoito/genética , Ratones , Ratones Endogámicos BALB C , Plasmodium falciparum/genética , Plasmodium yoelii/genética , Estructura Terciaria de Proteína , Conejos , Receptores de IgG/metabolismo , Linfocitos T/metabolismo , Linfocitos T/virología , Vacunas/genética
20.
J Antimicrob Chemother ; 67(10): 2501-5, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22763566

RESUMEN

OBJECTIVES: Recent work with the yeast model revealed that the antiprotozoal drug quinine competes with tryptophan for uptake via a common transport protein, causing cellular tryptophan starvation. In the present work, it was hypothesized that similar interactions may occur in malaria patients receiving quinine therapy. PATIENTS AND METHODS: A direct observational study was conducted in which plasma levels of drug and amino acids (tryptophan, tyrosine and phenylalanine) were monitored during quinine treatment of malaria patients with Plasmodium falciparum infections. RESULTS: Consistent with competition for uptake from plasma into cells, plasma tryptophan and tyrosine levels increased ≥2-fold during quinine therapy. Plasma quinine levels in individual plasma samples were significantly and positively correlated with tryptophan and tyrosine in the same samples. Control studies indicated no effect on phenylalanine. Chloroquine treatment of Plasmodium vivax-infected patients did not affect plasma tryptophan or tyrosine. During quinine treatment, plasma tryptophan was significantly lower (and quinine significantly higher) in patients experiencing adverse drug reactions. CONCLUSIONS: Plasma quinine levels during therapy are related to patient tryptophan and tyrosine levels, and these interactions can determine patient responses to quinine. The study also highlights the potential for extrapolating insights directly from the yeast model to human malaria patients.


Asunto(s)
Antimaláricos/administración & dosificación , Interacciones Farmacológicas , Malaria Falciparum/tratamiento farmacológico , Quinina/administración & dosificación , Triptófano/antagonistas & inhibidores , Tirosina/antagonistas & inhibidores , Adulto , Anciano , Antimaláricos/farmacología , Femenino , Humanos , Malaria Vivax/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Plasma/química , Quinina/farmacología , Triptófano/metabolismo , Tirosina/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA