Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Blood ; 119(17): 4073-82, 2012 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-22394599

RESUMEN

Today it is generally accepted that B cells require cognate interactions with CD4(+) T cells to develop high-affinity antibodies against proteins. CD4(+) T cells recognize peptides (epitopes) presented by MHC class II molecules that are expressed on antigen-presenting cells. Structural features of both the MHC class II molecule and the peptide determine the specificity of CD4(+) T cells that can bind to the MHC class II-peptide complex. We used a new humanized hemophilic mouse model to identify FVIII peptides presented by HLA-DRB1*1501. This model carries a knockout of all murine MHC class II molecules and expresses a chimeric murine-human MHC class II complex that contains the peptide-binding sites of the human HLA-DRB1*1501. When mice were treated with human FVIII, the proportion of mice that developed antibodies depended on the application route of FVIII and the activation state of the innate immune system. We identified 8 FVIII peptide regions that contained CD4(+) T-cell epitopes presented by HLA-DRB1*1501 to CD4(+) T cells during immune responses against FVIII. CD4(+) T-cell responses after intravenous and subcutaneous application of FVIII involved the same immunodominant FVIII epitopes. Interestingly, most of the 8 peptide regions contained promiscuous epitopes that bound to several different HLA-DR proteins in in vitro binding assays.


Asunto(s)
Formación de Anticuerpos/inmunología , Linfocitos T CD4-Positivos/inmunología , Modelos Animales de Enfermedad , Epítopos de Linfocito T/inmunología , Factor VIII/administración & dosificación , Factor VIII/inmunología , Cadenas HLA-DRB1/inmunología , Hemofilia A/inmunología , Animales , Presentación de Antígeno , Linfocitos T CD4-Positivos/metabolismo , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Cadenas HLA-DRB1/metabolismo , Haplotipos/genética , Hemofilia A/metabolismo , Hemofilia A/patología , Humanos , Inyecciones Intravenosas , Inyecciones Subcutáneas , Masculino , Ratones , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
2.
Blood ; 118(11): 3154-62, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21788339

RESUMEN

Memory B cells are involved in long-term maintenance of antibody-dependent immunologic disorders. Therefore, it is essential to understand how the restimulation of FVIII-specific memory B cells in hemophilia A with FVIII inhibitors is regulated. We asked whether concurrent activation of the innate immune system by an agonist for toll-like receptor (TLR) 7 is able to facilitate the differentiation of FVIII-specific memory B cells in the absence of T-cell help. TLR7 recognizes single-stranded RNA as contained in RNA viruses such as influenza, Sendai, and Coxsackie B viruses. Our results indicate that highly purified murine memory B cells do not differentiate into FVIII-specific antibody-secreting cells in the presence of FVIII and the TLR7 agonist when cultured in the absence of CD4(+) T cells. However, CD11c(+) dendritic cells facilitate the T cell-independent differentiation of FVIII-specific memory B cells but only in the presence of FVIII and the TLR7 agonist. In contrast to T cell-dependent restimulation, the antibody response after T cell-independent restimulation of FVIII-specific memory B cells is skewed toward IgG2a, an antibody subclass that is efficient in activating the complement system and in inducing Fc-receptor-mediated effector functions, both are required for effective immune responses against pathogens.


Asunto(s)
Linfocitos B/inmunología , Células Dendríticas/fisiología , Factor VIII/inmunología , Memoria Inmunológica/inmunología , Receptor Toll-Like 7/agonistas , Animales , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Células Cultivadas , Células Dendríticas/inmunología , Epítopos/efectos de los fármacos , Epítopos/inmunología , Memoria Inmunológica/efectos de los fármacos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Especificidad por Sustrato/efectos de los fármacos , Especificidad por Sustrato/inmunología , Linfocitos T/inmunología , Linfocitos T/fisiología
3.
Blood ; 118(13): 3698-707, 2011 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-21705497

RESUMEN

Replacement of the missing factor VIII (FVIII) is the current standard of care for patients with hemophilia A. However, the short half-life of FVIII makes frequent treatment necessary. Current efforts focus on the development of longer-acting FVIII concentrates by introducing chemical and genetic modifications to the protein. Any modification of the FVIII protein, however, risks increasing its immunogenic potential to induce neutralizing antibodies (FVIII inhibitors), and this is one of the major complications in current therapy. It would be highly desirable to identify candidates with a high risk for increased immunogenicity before entering clinical development to minimize the risk of exposing patients to such altered FVIII proteins. In the present study, we describe a transgenic mouse line that expresses a human F8 cDNA. This mouse is immunologically tolerant to therapeutic doses of native human FVIII but is able to mount an antibody response when challenged with a modified FVIII protein that possesses altered immunogenic properties. In this situation, immunologic tolerance breaks down and antibodies develop that recognize both the modified and the native human FVIII. The applicability of this new model for preclinical immunogenicity assessment of new FVIII molecules and its potential use for basic research are discussed.


Asunto(s)
Factor VIII/genética , Factor VIII/inmunología , Hemofilia A/genética , Tolerancia Inmunológica/genética , Memoria Inmunológica/genética , Animales , Formación de Anticuerpos/genética , Formación de Anticuerpos/fisiología , Modelos Animales de Enfermedad , Factor VIII/antagonistas & inhibidores , Femenino , Hemofilia A/inmunología , Hemofilia A/patología , Humanos , Tolerancia Inmunológica/fisiología , Memoria Inmunológica/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Especificidad de la Especie
4.
Pharm Res ; 30(11): 2855-67, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23775444

RESUMEN

PURPOSE: Human factor VIIa (FVIIa) is commonly used as bypassing therapy to treat bleeding episodes in hemophilia patients with neutralizing antibodies to factors VIII (FVIII) or IX (FIX). There is a need for a suitable animal model to assess the immunogenicity of new FVIIa products during preclinical development. The aim of this study was the design of a novel transgenic mouse model with immune tolerance to human FVIIa. METHODS: The model was generated by transgenic expression of human F7 cDNA. FVIIa-specific immune responses after treatment with human FVIIa were assessed by analyzing circulating antibodies, antibody producing plasma cells and CD4(+) T cells. RESULTS: In contrast to wild-type mice, human FVII transgenic mice did not develop antibodies when treated with human FVIIa. The immune tolerance was specific and could be broken by application of human FVIIa together with a strong stimulus of the innate immune system. Break of tolerance was associated with increased numbers of pro-inflammatory FVIIa-specific CD4(+) T cells. CONCLUSIONS: The new mouse model is suitable to study the influence of the innate immune system on maintenance and break of immune tolerance against FVIIa and could be used to assess the immunogenicity of new FVIIa products during pre-clinical development.


Asunto(s)
Factor VIIa/genética , Factor VIIa/inmunología , Tolerancia Inmunológica , Transgenes , Animales , Linfocitos T CD8-positivos/inmunología , ADN Complementario/genética , Factor VIIa/uso terapéutico , Femenino , Humanos , Inmunidad Innata , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Animales
5.
Drug Deliv Transl Res ; 13(9): 2367-2377, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-36920736

RESUMEN

Immunoglobulin replacement therapy is a life-saving treatment in patients with immunodeficiency and effective in the management of autoimmune disorders. Immunoglobulins are administered intravenously or subcutaneously, with the latter route reducing systemic reactions and providing an option for self-infusion, increasing patient convenience, while decreasing patient burden, healthcare utilization, and costs. A major limitation with subcutaneous administrations is the frequency of infusion due to limited volumes administrable into subcutaneous space, necessitating increased drug concentration, absorption, and dispersion. Increasing the concentration of immunoglobulins from 10 to 20% halves the required volume, but leads to higher dynamic viscosity, limiting infusion rate. Recombinant human hyaluronidase increases dispersion and absorption of immunoglobulins allowing administration of ≤ 600 mL per site, but does not change viscosity. Since the viscosity of fluids depends on temperature, we tested the feasibility of in-line warming of immunoglobulin formulations to physiological temperatures. In vitro analysis showed no negative impact of in-line warming to 38 °C on product quality. Subcutaneous infusion studies in pigs confirmed the feasibility of infusion rates of up to 7.5 mL/min with in-line warmed TAK-881, an immunoglobulin 20% facilitated with recombinant human hyaluronidase. In-line pressures were reduced compared with conventional immunoglobulin 20%, and local tolerance was not altered. Reduction of in-line pressures was more pronounced with thinner needle sets, indicating a potential benefit for patients. In summary, an in in-line warming device can circumvent the limitation of high viscosity, while product quality and local tolerance are maintained. The results of the presented studies warrant further testing in a phase 1 clinical study.


Asunto(s)
Hialuronoglucosaminidasa , Síndromes de Inmunodeficiencia , Humanos , Animales , Porcinos , Hialuronoglucosaminidasa/efectos adversos , Inmunoglobulinas/efectos adversos , Síndromes de Inmunodeficiencia/tratamiento farmacológico , Infusiones Subcutáneas , Inyecciones Subcutáneas
6.
Front Immunol ; 13: 975680, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36389737

RESUMEN

Background and aims: Hemophilia A is a severe bleeding disorder caused by the deficiency of functionally active coagulation factor VIII (FVIII). The induction of neutralizing anti-drug antibodies is a major complication in the treatment of hemophilia A patients with FVIII replacement therapies. Why some patients develop neutralizing antibodies (FVIII inhibitors) while others do not is not well understood. Previous studies indicated that the induction of FVIII inhibitors requires cognate interactions between FVIII-specific B cells and FVIII-specific CD4+ T cells in germinal center reactions. In this study, we investigated the FVIII peptide repertoire presented by antigen-presenting cells (APCs) under different microenvironment conditions that are expected to alter the uptake of FVIII by APCs. The aim of this study was to better understand the association between different microenvironment conditions during FVIII uptake and the FVIII peptide patterns presented by APCs. Methods: We used a FVIII-specific CD4+ T cell hybridoma library derived from humanized HLA-DRB1*1501 (human MHC class II) hemophilic mice that were treated with human FVIII. APCs obtained from the same mouse strain were preincubated with FVIII under different conditions which are expected to alter the uptake of FVIII by APCs. Subsequently, these preincubated APCs were used to stimulate the FVIII-specific CD4+ T cell hybridoma library. Stimulation of peptide-specific CD4+ T-cell hybridoma clones was assessed by analyzing the IL-2 release into cell culture supernatants. Results: The results of this study indicate that the specific microenvironment conditions during FVIII uptake by APCs determine the peptide specificities of subsequently activated FVIII-specific CD4+ T cell hybridoma clones. Incubation of APCs with FVIII complexed with von Willebrand Factor, FVIII activated by thrombin or FVIII combined with a blockade of receptors on APCs previously associated with FVIII uptake and clearance, resulted in distinct peptide repertoires of subsequently activated hybridoma clones. Conclusion: Based on our data we conclude that the specific microenvironment during FVIII uptake by APCs determines the FVIII peptide repertoire presented on MHC class II expressed by APCs and the peptide specificity of subsequently activated FVIII-specific CD4+ T cell hybridoma clones.


Asunto(s)
Hemofilia A , Hemostáticos , Humanos , Ratones , Animales , Factor VIII , Hemofilia A/tratamiento farmacológico , Linfocitos T CD4-Positivos , Células Presentadoras de Antígenos , Péptidos
7.
Proc Natl Acad Sci U S A ; 105(19): 6954-8, 2008 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-18458346

RESUMEN

Brown algae of the Laminariales (kelps) are the strongest accumulators of iodine among living organisms. They represent a major pump in the global biogeochemical cycle of iodine and, in particular, the major source of iodocarbons in the coastal atmosphere. Nevertheless, the chemical state and biological significance of accumulated iodine have remained unknown to this date. Using x-ray absorption spectroscopy, we show that the accumulated form is iodide, which readily scavenges a variety of reactive oxygen species (ROS). We propose here that its biological role is that of an inorganic antioxidant, the first to be described in a living system. Upon oxidative stress, iodide is effluxed. On the thallus surface and in the apoplast, iodide detoxifies both aqueous oxidants and ozone, the latter resulting in the release of high levels of molecular iodine and the consequent formation of hygroscopic iodine oxides leading to particles, which are precursors to cloud condensation nuclei. In a complementary set of experiments using a heterologous system, iodide was found to effectively scavenge ROS in human blood cells.


Asunto(s)
Antioxidantes/metabolismo , Atmósfera/química , Yoduros/metabolismo , Kelp/metabolismo , Depuradores de Radicales Libres/metabolismo , Ácidos Hexurónicos/farmacología , Laminaria/metabolismo , Modelos Biológicos , Ozono , Estallido Respiratorio , Análisis Espectral , Factores de Tiempo
8.
J Pharmacol Exp Ther ; 328(1): 107-15, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18829727

RESUMEN

5-Azacytidine (5-aza-CR) is a DNA-hypomethylating antineoplastic agent used because of its inhibitory activity on DNA methyltransferases. Today, it is approved as an epigenetically active drug therapy for treatment of myelodysplastic disorders, with a contraindication as to pre-existing liver diseases. Because the mechanism of its hepatotoxicity is still unknown, we investigated the pharmacodynamic properties of 5-aza-CR with regard to death receptor/ligand-induced apoptosis and the mode of execution of cell death. In a time- and concentration-dependent manner, primary murine, human hepatocytes and HepG2 cells exposed to 5-aza-CR became highly sensitive toward cell death induced by CD95L, tumor necrosis factor (TNF)-related apoptosis-inducing ligand, or TNF. Cell death was characterized as apoptotic by membrane blebbing, chromatin condensation, and exposure of phosphatidylserine on the outer membrane. Neither 5-aza-2'-deoxycytidine nor the common DNA methyltransferase inhibitors S-(5'-adenosyl)-L-homocysteine or RG 108 showed any significant effects under these conditions. Despite the complete protection of HepG2 by high concentrations of the pan-caspase inhibitor N-benzyloxycarbonyl-Val-Ala-Asp(O-Me) fluoromethyl ketone (z-VAD-fmk), effector caspase-3/7 activity was completely abolished at approximately a 20-fold lower concentration of z-VAD-fmk. Under these conditions, the serine protease inhibitors N,alpha-tosyl-L-phenylalanine chloromethyl ketone, N,p-tosyl-L-lysine chloromethyl ketone, and 4-(2-aminoethyl)-benzenesulfonyl fluoride, respectively, conferred protection against death receptor ligands. We conclude that this caspase-independent apoptosis is executed by a yet-unidentified serine protease.


Asunto(s)
Apoptosis/fisiología , Azacitidina/farmacología , Hepatocitos/fisiología , Hígado/citología , Hígado/fisiología , Receptores de Muerte Celular/agonistas , Receptores de Muerte Celular/fisiología , Animales , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Técnicas de Cultivo de Célula , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Humanos , Hígado/efectos de los fármacos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/cirugía , Ratones , Microscopía Fluorescente
9.
Toxicology ; 201(1-3): 21-30, 2004 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15297016

RESUMEN

The essential trace element selenium is also toxic at low doses. Since supplementation of selenium is discussed as cancer prophylaxis, we investigated whether or not bioavailable selenium compounds are selectively toxic on malignant cells by comparing primary and transformed liver cells as to the extent and mode of cell death. Sodium selenite and selenate exclusively induced necrosis in a concentration-dependent manner in all cell types investigated. In primary murine hepatocytes, the EC50 was 20 microM for selenite, 270 microM for selenate, and 30 microM for Se-methionine. In the human carcinoma cell line HepG2, the EC50 for selenite was 40 microM, and for selenate 1.1 mM, whereas Se-methionine was essentially non-toxic up to 10 mM. Similar results were found in murine Hepa1-6 cells. Exposure of primary murine cells to selenate or selenite resulted in increased lipid peroxidation. Toxicity was inhibited by superoxide dismutase plus catalase, indicating an important role for reactive oxygen intermediates. In primary hepatocytes, metabolical depletion of intracellular ATP by the ketohexose tagatose, significantly decreased the cytotoxicity of Se-methionine, while the one of selenite was increased. These data do not provide any in vitro evidence that bioavailable selenium compounds induce preferentially apoptotic cell death or selectively kill transformed hepatocytes.


Asunto(s)
Apoptosis/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Selenio/toxicidad , Animales , Disponibilidad Biológica , Caspasa 3 , Caspasas/efectos de los fármacos , Caspasas/metabolismo , Línea Celular Tumoral , Células Cultivadas , Hepatocitos/enzimología , Hepatocitos/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Ratas , Selenio/metabolismo
10.
Exp Toxicol Pathol ; 63(5): 511-7, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20537879

RESUMEN

Histone deacetylases (HD) represent a novel target in cancer treatment, particularly for scattered small tumours such as the hepatocellular carcinoma (HCC). However, only few studies address the toxicological impact of HD Inhibitors (HDIs) on malignantly transformed cells versus primary hepatocytes. We examined whether and how different classes of HDIs sensitise the human HCC cell line HepG2, primary healthy murine and human liver cells towards the death receptor agonists TNFα and CD95L. Apicidin, M344 (N-hydroxy-7-(-4-dimethylaminobenzol)aminoheptanamide), CBHA (m-carboxycinnamic acid bis-hydroxamide) and VPA (valproic acid) sensitised liver cell cultures towards CD95-triggered apoptosis with the following potency: apicidin > M344 ≈ CBHA ≫ VPA. Apicidin sensitised towards CD95 also in the intact organ, i.e. in the isolated perfused mouse liver. No significant sensitisation towards TNFα was found in vitro. Western blot analysis showed that all HDIs studied downregulated the anti-apoptotic protein cFLIP, but only VPA additionally affected the expression level of XIAP. Furthermore, in models of the intrinsic apoptosis pathway, i.e. in HepG2 cells treated with Melphalan and in primary hepatocytes irradiated with UV light, only VPA exhibited significant sensitisation. These findings extend the biochemical, pharmacological and toxicological basis for HDI therapy and provide a caveat for clinical use in patients with an accompanying critical inflammatory state in which the CD95 system might be pre-activated.


Asunto(s)
Antineoplásicos/inmunología , Antineoplásicos/toxicidad , Hepatocitos/efectos de los fármacos , Inhibidores de Histona Desacetilasas/inmunología , Inhibidores de Histona Desacetilasas/toxicidad , Animales , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Western Blotting , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Caspasa 8/genética , Caspasa 8/metabolismo , Técnicas de Cultivo de Célula , Supervivencia Celular/efectos de los fármacos , Proteína Ligando Fas/inmunología , Proteína Ligando Fas/metabolismo , Células Hep G2 , Hepatocitos/enzimología , Hepatocitos/inmunología , Histona Desacetilasas/metabolismo , Humanos , Hígado/efectos de los fármacos , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Perfusión , Transfección , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA