Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Cytometry A ; 105(2): 124-138, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37751141

RESUMEN

Flow cytometry is the method of choice for immunophenotyping in the context of clinical, translational, and systems immunology studies. Among the latter, the Milieu Intérieur (MI) project aims at defining the boundaries of a healthy immune response to identify determinants of immune response variation. MI used immunophenotyping of a 1000 healthy donor cohort by flow cytometry as a principal outcome for immune variance at steady state. New generation spectral cytometers now enable high-dimensional immune cell characterization from small sample volumes. Therefore, for the MI 10-year follow up study, we have developed two high-dimensional spectral flow cytometry panels for deep characterization of innate and adaptive whole blood immune cells (35 and 34 fluorescent markers, respectively). We have standardized the protocol for sample handling, staining, acquisition, and data analysis. This approach enables the reproducible quantification of over 182 immune cell phenotypes at a single site. We have applied the protocol to discern minor differences between healthy and patient samples and validated its value for application in immunomonitoring studies. Our protocol is currently used for characterization of the impact of age and environmental factors on peripheral blood immune phenotypes of >400 donors from the initial MI cohort.


Asunto(s)
Estudios de Seguimiento , Humanos , Inmunofenotipificación , Fenotipo , Citometría de Flujo/métodos
2.
J Immunoassay Immunochem ; 43(5): 467-479, 2022 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-35301912

RESUMEN

Antigens derived from engulfed apoptotic bodies that are presented by dendritic cells can amplify Ag-specific T-cells. Accelerated co-cultured DC (acDC) strategy keeps lymphocytes in contact with differentiating DCs. Therefore, Ag-specific T-cell activation can occur during DC maturation. Our aim was to prepare DCs by acDC method and check the subsequent engulfment of the apoptotic body by acDC. We have proposed that this method could be feasible if we transfect the apoptotic bodies with the antigen. DCs were prepared using acDC method and their maturation markers were confirmed by flow cytometry. Ultraviolet was used for inducing apoptosis in the PBMCs and induction of apoptosis checked by propidium iodide and 7-aminoactinomycin D staining. Flow cytometry and immunohistochemistry were used for checking the uptake of apoptotic bodies by the DCs. The alloreactivity against apoptotic bodies was examined by enzyme-linked immunospot (ELISPOT) assay. Results showed that 40.4% of DCs could efficiently engulf the apoptotic bodies. The results indicated that acDC method is capable to isolate a high yield of DCs, and these cells could properly engulf the apoptotic bodies, more works should be performed to use this method for Ag discovery through delivering the Ag by apoptotic bodies into the DCs.


Asunto(s)
Células Dendríticas , Vesículas Extracelulares , Antígenos , Apoptosis , Activación de Linfocitos , Linfocitos T
3.
Diabetologia ; 61(3): 658-670, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29196783

RESUMEN

AIMS/HYPOTHESIS: Validated biomarkers are needed to monitor the effects of immune intervention in individuals with type 1 diabetes. Despite their importance, few options exist for monitoring antigen-specific T cells. Previous reports described a combinatorial approach that enables the simultaneous detection and quantification of multiple islet-specific CD8+ T cell populations. Here, we set out to evaluate the performance of a combinatorial HLA-A2 multimer assay in a multi-centre setting. METHODS: The combinatorial HLA-A2 multimer assay was applied in five participating centres using centralised reagents and blinded replicate samples. In preliminary experiments, samples from healthy donors were analysed using recall antigen multimers. In subsequent experiments, samples from healthy donors and individuals with type 1 diabetes were analysed using beta cell antigen and recall antigen multimers. RESULTS: The combinatorial assay was successfully implemented in each participating centre, with CVs between replicate samples that indicated good reproducibility for viral epitopes (mean %CV = 33.8). For beta cell epitopes, the assay was very effective in a single-centre setting (mean %CV = 18.4), but showed sixfold greater variability across multi-centre replicates (mean %CV = 119). In general, beta cell antigen-specific CD8+ T cells were detected more commonly in individuals with type 1 diabetes than in healthy donors. Furthermore, CD8+ T cells recognising HLA-A2-restricted insulin and glutamate decarboxylase epitopes were found to occur at higher frequencies in individuals with type 1 diabetes than in healthy donors. CONCLUSIONS/INTERPRETATION: Our results suggest that, although combinatorial multimer assays are challenging, they can be implemented in multiple laboratories, providing relevant T cell frequency measurements. Assay reproducibility was notably higher in the single-centre setting, suggesting that biomarker analysis of clinical trial samples would be most successful when assays are performed in a single laboratory. Technical improvements, including further standardisation of cytometry platforms, will likely be necessary to reduce assay variability in the multi-centre setting.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Antígeno HLA-A2/metabolismo , Adulto , Biomarcadores/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Femenino , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Masculino , Adulto Joven
4.
J Immunol ; 192(11): 4957-66, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24778449

RESUMEN

Peptide ligands presented by MHC class I (MHC-I) molecules are produced by degradation of cytosolic and nuclear, but also endoplasmic reticulum (ER)-resident, proteins by the proteasome. However, Ag processing of ER proteins remains little characterized. Studying processing and presentation of proinsulin, which plays a pivotal role in autoimmune diabetes, we found that targeting to the ER has profound effects not only on how proinsulin is degraded, but also on regulation of its cellular levels. While proteasome inhibition inhibited degradation and presentation of cytosolic proinsulin, as expected, it reduced the abundance of ER-targeted proinsulin. This targeting and protein modifications modifying protein half-life also had profound effects on MHC-I presentation and proteolytic processing of proinsulin. Thus, presentation of stable luminal forms was inefficient but enhanced by proteasome inhibition, whereas that of unstable luminal forms and of a cytosolic form were more efficient and compromised by proteasome inhibitors. Distinct stability of peptide MHC complexes produced from cytosolic and luminal proinsulin suggests that different proteolytic activities process the two Ag forms. Thus, both structural features and subcellular targeting of Ags can have strong effects on the processing pathways engaged by MHC-I-restricted Ags, and on the efficiency and regulation of their presentation.


Asunto(s)
Presentación de Antígeno , Retículo Endoplásmico/inmunología , Regulación de la Expresión Génica/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Proinsulina/inmunología , Proteolisis , Retículo Endoplásmico/genética , Regulación de la Expresión Génica/genética , Células HeLa , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Péptidos/genética , Péptidos/inmunología , Proinsulina/genética
5.
Eur J Immunol ; 42(6): 1553-61, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22678909

RESUMEN

Despite encouraging results in the NOD mouse, type 1 diabetes prevention trials using subcutaneous insulin have been unsuccessful. To explain these discrepancies, 3-week-old NOD mice were treated for 7 weeks with subcutaneous insulin at two different doses: a high dose (0.5 U/mouse) used in previous mouse studies; and a low dose (0.005 U/mouse) equivalent to that used in human trials. Effects on insulitis and diabetes were monitored along with immune and metabolic modifications. Low-dose insulin did not have any effect on disease incidence. High-dose treatment delayed but did not prevent diabetes, with reduced insulitis reappearing once insulin discontinued. This effect was not associated with significant immune changes in islet infiltrates, either in terms of cell composition or frequency and IFN-γ secretion of islet-reactive CD8(+) T cells recognizing the immunodominant epitopes insulin B(15-23) and islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP)(206-214). Delayed diabetes and insulitis were associated with lower blood glucose and endogenous C-peptide levels, which rapidly returned to normal upon treatment discontinuation. In conclusion, high- but not low-dose prophylactic insulin treatment delays diabetes onset and is associated with metabolic changes suggestive of ß-cell "rest" which do not persist beyond treatment. These findings have important implications for designing insulin-based prevention trials.


Asunto(s)
Diabetes Mellitus Tipo 1/prevención & control , Hipoglucemiantes/administración & dosificación , Insulina/administración & dosificación , Animales , Glucemia/análisis , Péptido C/metabolismo , Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Inyecciones Subcutáneas , Interferón gamma/biosíntesis , Ratones , Ratones Endogámicos NOD
6.
Curr Diab Rep ; 11(5): 426-33, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21732231

RESUMEN

The central role of T cells in type 1 diabetes pathogenesis is well established, but these cells continue to pose numerous challenges in understanding their dynamics and in following their modifications. Important progress has been recently made in pinpointing some novel antigens targeted by pathogenic T cells and the epitope sequences recognized. Studies on the interplay between effector T cells, their regulatory counterparts, and cells of the innate immune system have unraveled novel pathways and may inspire new therapeutic approaches. At the same time, the appreciation of the plasticity of regulatory T cells has raised important caveats on their use for cell-based therapies. Continuous development of T-cell assays exploring both pathogenic and regulatory players will be critical to "take the temperature" of undergoing disease progression and reversal.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Linfocitos T Reguladores/inmunología , Animales , Epítopos/inmunología , Humanos , Inmunidad Innata/inmunología
7.
Clin Dev Immunol ; 2011: 286248, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21647401

RESUMEN

The ideal drug of modern medicine is the one that achieves its therapeutic target with minimal adverse effects. Immune therapy of Type 1 diabetes (T1D) is no exception, and knowledge of the antigens targeted by pathogenic T cells offers a unique opportunity towards this goal. Different antigen formulations are being considered, such as proteins or peptides, either in their native form or modified ad hoc, DNA plasmids, and cell-based agents. Translation from mouse to human should take into account important differences, particularly in the time scale of autoimmune progression, and intervention. Critical parameters such as administration route, dosing and interval remain largely empirical and need to be further dissected. T1D staging through immune surrogate markers before and after treatment will be key in understanding therapeutic actions and to finally turn ordinary blanks into magic bullets.


Asunto(s)
Antígenos/inmunología , Diabetes Mellitus Tipo 1/terapia , Inmunoterapia , Animales , Antígenos/uso terapéutico , Biomarcadores/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Humanos , Ratones , Ratones Endogámicos NOD , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo
8.
Front Immunol ; 12: 616215, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34447366

RESUMEN

Tolerogenic vaccinations using beta-cell antigens are attractive for type 1 diabetes prevention, but clinical trials have been disappointing. This is probably due to the late timing of intervention, when multiple auto-antibodies are already present. We therefore devised a strategy to introduce the initiating antigen preproinsulin (PPI) during neonatal life, when autoimmunity is still silent and central tolerance mechanisms, which remain therapeutically unexploited, are more active. This strategy employs an oral administration of PPI-Fc, i.e. PPI fused with an IgG Fc to bind the intestinal neonatal Fc receptor (FcRn) that physiologically delivers maternal antibodies to the offspring during breastfeeding. Neonatal oral PPI-Fc vaccination did not prevent diabetes development in PPI T-cell receptor-transgenic G9C8.NOD mice. However, PPI-Fc was efficiently transferred through the intestinal epithelium in an Fc- and FcRn-dependent manner, was taken up by antigen presenting cells, and reached the spleen and thymus. Although not statistically significant, neonatal oral PPI-Fc vaccination delayed diabetes onset in polyclonal Ins2-/-.NOD mice that spontaneously develop accelerated diabetes. Thus, this strategy shows promise in terms of systemic and thymic antigen delivery via the intestinal FcRn pathway, but the current PPI-Fc formulation/regimen requires further improvements to achieve diabetes prevention.


Asunto(s)
Diabetes Mellitus Experimental/prevención & control , Diabetes Mellitus Tipo 1/prevención & control , Antígenos de Histocompatibilidad Clase I/inmunología , Insulina/farmacología , Precursores de Proteínas/farmacología , Receptores Fc/inmunología , Proteínas Recombinantes de Fusión/farmacología , Timo/inmunología , Administración Oral , Animales , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Insulina/genética , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Precursores de Proteínas/genética , Receptores Fc/genética , Proteínas Recombinantes de Fusión/genética
9.
Sci Immunol ; 3(20)2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29429978

RESUMEN

The human leukocyte antigen-A2 (HLA-A2)-restricted zinc transporter 8186-194 (ZnT8186-194) and other islet epitopes elicit interferon-γ secretion by CD8+ T cells preferentially in type 1 diabetes (T1D) patients compared with controls. We show that clonal ZnT8186-194-reactive CD8+ T cells express private T cell receptors and display equivalent functional properties in T1D and healthy individuals. Ex vivo analyses further revealed that CD8+ T cells reactive to ZnT8186-194 and other islet epitopes circulate at similar frequencies and exhibit a predominantly naïve phenotype in age-matched T1D and healthy donors. Higher frequencies of ZnT8186-194-reactive CD8+ T cells with a more antigen-experienced phenotype were detected in children versus adults, irrespective of disease status. Moreover, some ZnT8186-194-reactive CD8+ T cell clonotypes were found to cross-recognize a Bacteroides stercoris mimotope. Whereas ZnT8 was poorly expressed in thymic medullary epithelial cells, variable thymic expression levels of islet antigens did not modulate the peripheral frequency of their cognate CD8+ T cells. In contrast, ZnT8186-194-reactive cells were enriched in the pancreata of T1D patients versus nondiabetic and type 2 diabetic individuals. Thus, islet-reactive CD8+ T cells circulate in most individuals but home to the pancreas preferentially in T1D patients. We conclude that the activation of this common islet-reactive T cell repertoire and progression to T1D likely require defective peripheral immunoregulation and/or a proinflammatory islet microenvironment.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Islotes Pancreáticos/inmunología , Páncreas/citología , Páncreas/inmunología , Adulto , Línea Celular , Niño , Femenino , Antígeno HLA-A2/inmunología , Voluntarios Sanos , Humanos , Masculino
10.
Cell Metab ; 28(6): 946-960.e6, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30078552

RESUMEN

Although CD8+ T-cell-mediated autoimmune ß cell destruction occurs in type 1 diabetes (T1D), the target epitopes processed and presented by ß cells are unknown. To identify them, we combined peptidomics and transcriptomics strategies. Inflammatory cytokines increased peptide presentation in vitro, paralleling upregulation of human leukocyte antigen (HLA) class I expression. Peptide sources featured several insulin granule proteins and all known ß cell antigens, barring islet-specific glucose-6-phosphatase catalytic subunit-related protein. Preproinsulin yielded HLA-A2-restricted epitopes previously described. Secretogranin V and its mRNA splice isoform SCG5-009, proconvertase-2, urocortin-3, the insulin gene enhancer protein ISL-1, and an islet amyloid polypeptide transpeptidation product emerged as antigens processed into HLA-A2-restricted epitopes, which, as those already described, were recognized by circulating naive CD8+ T cells in T1D and healthy donors and by pancreas-infiltrating cells in T1D donors. This peptidome opens new avenues to understand antigen processing by ß cells and for the development of T cell biomarkers and tolerogenic vaccination strategies.


Asunto(s)
Presentación de Antígeno , Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Epítopos de Linfocito T/inmunología , Transcriptoma/inmunología , Animales , Biomarcadores/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Estudios de Casos y Controles , Línea Celular , Hormona Liberadora de Corticotropina/metabolismo , Citocinas/metabolismo , Antígenos HLA/metabolismo , Humanos , Insulina/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Ratones , Proteína 7B2 Secretora Neuroendocrina/metabolismo , Proproteína Convertasa 2/metabolismo , Precursores de Proteínas/metabolismo , Proteómica/métodos , Urocortinas/metabolismo
11.
Sci Rep ; 7(1): 4122, 2017 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-28646234

RESUMEN

In vivo electroporation (EP) is used to enhance the uptake of nucleic acids and its association with DNA vaccination greatly stimulates immune responses to vaccine antigens delivered through the skin. However, the effect of EP on cutaneous cell behavior, the dynamics of immune cell recruitment and local inflammatory factors, have not been fully described. Here, we show that intradermal DNA vaccination combined with EP extends antigen expression to the epidermis and the subcutaneous skin muscle in non-human primates. In vivo fibered confocal microscopy and dynamic ex vivo imaging revealed that EP promotes the mobility of Langerhans cells (LC) and their interactions with transfected cells prior to their migration from the epidermis. At the peak of vaccine expression, we detected antigen in damaged keratinocyte areas in the epidermis and we characterized recruited immune cells in the skin, the hypodermis and the subcutaneous muscle. EP alone was sufficient to induce the production of pro-inflammatory cytokines in the skin and significantly increased local concentrations of Transforming Growth Factor (TGF)-alpha and IL-12. Our results show the kinetics of inflammatory processes in response to EP of the skin, and reveal its potential as a vaccine adjuvant.


Asunto(s)
Adyuvantes Inmunológicos , Electroporación , Inmunización , Plásmidos/administración & dosificación , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígenos/genética , Antígenos/inmunología , Citocinas/metabolismo , Epidermis/inmunología , Epidermis/metabolismo , Expresión Génica , Inmunización/métodos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Inyecciones Intradérmicas , Queratinocitos/inmunología , Queratinocitos/metabolismo , Células de Langerhans/inmunología , Células de Langerhans/metabolismo , Macaca , Plásmidos/genética , Vacunación/métodos , Vacunas de ADN/genética
12.
Diabetes ; 64(10): 3532-42, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25918233

RESUMEN

The first signs of autoimmune activation leading to ß-cell destruction in type 1 diabetes (T1D) appear during the first months of life. Thus, the perinatal period offers a suitable time window for disease prevention. Moreover, thymic selection of autoreactive T cells is most active during this period, providing a therapeutic opportunity not exploited to date. We therefore devised a strategy by which the T1D-triggering antigen preproinsulin fused with the immunoglobulin (Ig)G Fc fragment (PPI-Fc) is delivered to fetuses through the neonatal Fc receptor (FcRn) pathway, which physiologically transfers maternal IgGs through the placenta. PPI-Fc administered to pregnant PPIB15-23 T-cell receptor-transgenic mice efficiently accumulated in fetuses through the placental FcRn and protected them from subsequent diabetes development. Protection relied on ferrying of PPI-Fc to the thymus by migratory dendritic cells and resulted in a rise in thymic-derived CD4(+) regulatory T cells expressing transforming growth factor-ß and in increased effector CD8(+) T cells displaying impaired cytotoxicity. Moreover, polyclonal splenocytes from nonobese diabetic (NOD) mice transplacentally treated with PPI-Fc were less diabetogenic upon transfer into NOD.scid recipients. Transplacental antigen vaccination provides a novel strategy for early T1D prevention and, further, is applicable to other immune-mediated conditions.


Asunto(s)
Diabetes Mellitus Tipo 1/prevención & control , Antígenos de Histocompatibilidad Clase I/metabolismo , Insulina/metabolismo , Intercambio Materno-Fetal/fisiología , Precursores de Proteínas/metabolismo , Receptores Fc/metabolismo , Animales , Autoinmunidad , Proliferación Celular , Células Dendríticas/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Insulina/administración & dosificación , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Placenta/metabolismo , Embarazo , Precursores de Proteínas/administración & dosificación , Receptores Fc/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Organismos Libres de Patógenos Específicos , Timo/fisiología
13.
Sci Transl Med ; 7(275): 275ra21, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-25696000

RESUMEN

Central tolerance plays a key role in modulating immune responses to self and exogenous antigens. The absence of self-antigen expression, as in patients with genetic deficiencies, prevents the development of antigen-specific immune tolerance. Hence, a substantial number of patients develop neutralizing antibodies to the corresponding protein therapeutics after replacement treatment. In this context, the administration of missing antigens during fetal development, a key period for self-tolerance establishment, should confer early and long-lasting antigen-specific tolerance. To this end, we exploited the physiological pathway of the neonatal Fc receptor (FcRn) through which maternal immunoglobulins are transplacentally transferred to fetuses. We demonstrate that Fc-fused antigens administered to pregnant mice reach fetal lymphoid organs in an FcRn-dependent manner, accumulate in antigen-presenting cells of myeloid origin, and promote the generation of both thymic and peripheral antigen-specific regulatory T cells. This strategy was successfully pursued in a mouse model of hemophilia A, where maternofetal transfer of the Fc-fused immunodominant domains of coagulation factor VIII conferred antigen-specific tolerance. Transplacental tolerance induction with Fc-fused proteins may thus prove valuable to prevent alloimmunization after replacement protein therapy for congenital deficiencies.


Asunto(s)
Factor VIII/uso terapéutico , Tolerancia Inmunológica , Placenta/inmunología , Linfocitos T/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Células Presentadoras de Antígenos/inmunología , Endocitosis , Factor VIII/inmunología , Femenino , Hemofilia A/terapia , Intercambio Materno-Fetal , Ratones , Embarazo
14.
PLoS One ; 9(2): e88365, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24516642

RESUMEN

Antigen presentation by MHC class I molecules requires degradation of epitope source proteins in the cytosol. Although the preeminent role of the proteasome is clearly established, evidence suggesting a significant role for proteasome-independent generation of class I ligands has been reported repeatedly. However, an enzyme responsible for such a role has not been identified. Recently insulin-degrading enzyme (IDE) was shown to produce an antigenic peptide derived from the tumor antigen MAGE-A3 in an entirely proteasome-independent manner, raising the question of the global impact of IDE in MHC class I antigen processing. Here we report that IDE knockdown in human cell lines, or knockout in two different mouse strains, has no effect on cell surface expression of various MHC class I molecules, including allomorphs such as HLA-A3 and HLA-B27 suggested to be loaded in an at least a partly proteasome-independent manner. Moreover, reduced or absent IDE expression does not affect presentation of five epitopes including epitopes derived from beta amyloid and proinsulin, two preferred IDE substrates. Thus, IDE does not play a major role in MHC class I antigen processing, confirming the dominant and almost exclusive role of the proteasome in cytosolic production of MHC class I ligands.


Asunto(s)
Presentación de Antígeno/inmunología , Genes MHC Clase I/inmunología , Insulisina/metabolismo , Animales , Línea Celular Tumoral , Citosol/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Insulisina/genética , Ratones , Ratones Noqueados , Complejo de la Endopetidasa Proteasomal/metabolismo
15.
PLoS One ; 8(6): e67705, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23826334

RESUMEN

Insulin Degrading Enzyme (IDE) is a protease conserved through evolution with a role in diabetes and Alzheimer's disease. The reason underlying its ubiquitous expression including cells lacking identified IDE substrates remains unknown. Here we show that the fission yeast IDE homologue (Iph1) modulates cellular sensitivity to endoplasmic reticulum (ER) stress in a manner dependent on TORC1 (Target of Rapamycin Complex 1). Reduced sensitivity to tunicamycin was associated with a smaller number of cells undergoing apoptosis. Wild type levels of tunicamycin sensitivity were restored in iph1 null cells when the TORC1 complex was inhibited by rapamycin or by heat inactivation of the Tor2 kinase. Although Iph1 cleaved hallmark IDE substrates including insulin efficiently, its role in the ER stress response was independent of its catalytic activity since expression of inactive Iph1 restored normal sensitivity. Importantly, wild type as well as inactive human IDE complemented gene-invalidated yeast cells when expressed at the genomic locus under the control of iph1(+) promoter. These results suggest that IDE has a previously unknown function unrelated to substrate cleavage, which links sensitivity to ER stress to a pro-survival role of the TORC1 pathway.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de los fármacos , Eliminación de Gen , Insulisina/química , Complejos Multiproteicos/metabolismo , Schizosaccharomyces/enzimología , Serina-Treonina Quinasas TOR/metabolismo , Secuencia de Aminoácidos , Apoptosis/efectos de los fármacos , Citoprotección/efectos de los fármacos , Ditiotreitol/farmacología , Endopeptidasas/química , Endopeptidasas/metabolismo , Prueba de Complementación Genética , Genoma Fúngico/genética , Humanos , Insulina/química , Insulina/metabolismo , Insulisina/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Modelos Moleculares , Datos de Secuencia Molecular , Schizosaccharomyces/genética , Proteínas de Schizosaccharomyces pombe/química , Proteínas de Schizosaccharomyces pombe/metabolismo , Alineación de Secuencia , Sirolimus/farmacología , Tunicamicina/farmacología
16.
PLoS One ; 6(12): e28866, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22194932

RESUMEN

Synthetic peptides are widely used in immunological research as epitopes to stimulate their cognate T cells. These preparations are never completely pure, but trace contaminants are commonly revealed by mass spectrometry quality controls. In an effort to characterize novel major histocompatibility complex (MHC) Class I-restricted ß-cell epitopes in non-obese diabetic (NOD) mice, we identified islet-infiltrating CD8+ T cells recognizing a contaminating peptide. The amount of this contaminant was so small to be undetectable by direct mass spectrometry. Only after concentration by liquid chromatography, we observed a mass peak corresponding to an immunodominant islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP)(206-214) epitope described in the literature. Generation of CD8+ T-cell clones recognizing IGRP(206-214) using a novel method confirmed the identity of the contaminant, further underlining the immunodominance of IGRP(206-214). If left undetected, minute impurities in synthetic peptide preparations may thus give spurious results.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Espectrometría de Masas/métodos , Péptidos/inmunología , Secuencia de Aminoácidos , Animales , Células Clonales , Epítopos de Linfocito T/inmunología , Glucosa-6-Fosfatasa/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Interferón gamma/inmunología , Islotes Pancreáticos/inmunología , Ratones , Ratones Endogámicos NOD , Datos de Secuencia Molecular , Péptidos/química , Proteínas/inmunología , Reproducibilidad de los Resultados , Análisis de Secuencia de Proteína
18.
Hum Gene Ther ; 20(11): 1291-307, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19627235

RESUMEN

Strategies to improve vaccine efficacy are still required, especially in the case of chronic infections, including human immunodeficiency virus (HIV). DNA vaccines have potential advantages over conventional vaccines; however, low immunological efficacy has been demonstrated in many experiments involving large animals and in clinical trials. To improve the immunogenicity of DNA vaccines, we have designed a plasmid vector exploiting the binding capacity of the bovine papillomavirus E2 protein and we have used electroporation (EP) to increase DNA uptake after intradermal inoculation. We demonstrated, in nonhuman primates (NHPs), efficient induction of anti-HIV immunity with an improved DNA vaccine vector encoding an artificial fusion protein, consisting of several proteins and selected epitopes from HIV-1. We show that a DNA vaccine delivery method combining intradermal injection and noninvasive EP dramatically increased expression of the vaccine antigen selectively in the epidermis, and our observations strongly suggest the involvement of Langerhans cells in the strength and quality of the anti-HIV immune response. Although the humoral responses to the vaccine were transient, the cellular responses were exceptionally robust and persisted, at high levels, more than 2 years after the last vaccine boost. The immune responses were characterized by the induction of significant proportions of T cells producing both interferon-gamma and interleukin-2 cytokines, in both subpopulations, CD4(+) and CD8(+). This strategy is an attractive approach for vaccination in humans because of its high efficacy and the possible use of newly developed devices for EP.


Asunto(s)
Vacunas contra el SIDA/efectos adversos , Vectores Genéticos/inmunología , Infecciones por VIH/prevención & control , VIH-1/inmunología , Piel/inmunología , Animales , Citocinas/inmunología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Electroporación , Ensayo de Inmunoadsorción Enzimática , Epítopos de Linfocito T/genética , Vectores Genéticos/genética , Inmunohistoquímica , Células de Langerhans/inmunología , Macaca fascicularis , Estadísticas no Paramétricas , Linfocitos T/inmunología , Resultado del Tratamiento , Proteínas Virales/genética , Proteínas Virales/metabolismo
19.
J Biol Chem ; 279(52): 54210-5, 2004 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-15494401

RESUMEN

The assembly of MHC class I molecules with beta(2)-microglobulin and peptides is assisted by the housekeeping chaperones calnexin, calreticulin, and Erp57 and the dedicated accessory protein, tapasin. Tapasin and calreticulin are essential for efficient MHC class I assembly, but their precise action during class I assembly remains to be elucidated. Previous in vitro studies have demonstrated that the lectin calreticulin interacts with monoglucosylated MHC class I heavy chains, whatever their state of assembly with light chains and peptide, and inhibits their aggregation above physiological temperature. We used a soluble single chain HLA-A2/beta(2)-microglobulin molecule, A2SC, to study the effect of calreticulin on the peptide binding capacity of HLA class I molecules. Calreticulin inhibited the formation of A2SC aggregates both when co-expressed in insect cells and during incubations at elevated temperature. Calreticulin dramatically enhanced acquisition of peptide binding capacity when added to denatured A2SC molecules during refolding at 4 degrees C. However, it had no effect on the rapid loss of A2SC peptide binding capacity at physiological temperature. We conclude that calreticulin promotes the folding of HLA class I molecules to a state in which, at low temperature, they spontaneously acquire peptide binding capacity. However, it does not induce or maintain a peptide-receptive state of the class I-binding site, which is likely to be promoted by one or several other components of the class I loading complexes. By being amenable to complementation with additional proteins, the described system should be useful for identification of these components.


Asunto(s)
Calreticulina/fisiología , Antígenos de Histocompatibilidad Clase I/química , Animales , Antiportadores/genética , Antiportadores/fisiología , Baculoviridae/genética , Sitios de Unión , Calnexina/genética , Calnexina/fisiología , Calreticulina/genética , Calreticulina/farmacología , Línea Celular , Estabilidad de Medicamentos , Expresión Génica , Antígeno HLA-A2/química , Antígeno HLA-A2/genética , Antígeno HLA-A2/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Calor , Humanos , Inmunoglobulinas/genética , Inmunoglobulinas/fisiología , Proteínas de Transporte de Membrana , Unión Proteica/efectos de los fármacos , Pliegue de Proteína , Proteínas Recombinantes de Fusión , Spodoptera/metabolismo , Transfección , Microglobulina beta-2/química , Microglobulina beta-2/genética , Microglobulina beta-2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA