Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Diabetologia ; 65(10): 1701-1709, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35867130

RESUMEN

AIMS/HYPOTHESIS: Enteroviral infection has been implicated consistently as a key environmental factor correlating with the appearance of autoimmunity and/or the presence of overt type 1 diabetes, in which pancreatic insulin-producing beta cells are destroyed by an autoimmune response. Genetic predisposition through variation in the type 1 diabetes risk gene IFIH1 (interferon induced with helicase C domain 1), which encodes the viral pattern-recognition receptor melanoma differentiation-associated protein 5 (MDA5), supports a potential link between enterovirus infection and type 1 diabetes. METHODS: We used molecular techniques to detect enterovirus RNA in peripheral blood samples (in separated cellular compartments or plasma) from two cohorts comprising 79 children or 72 adults that include individuals with and without type 1 diabetes who had multiple autoantibodies. We also used immunohistochemistry to detect the enteroviral protein VP1 in the pancreatic islets of post-mortem donors (n=43) with type 1 diabetes. RESULTS: We observed enhanced detection sensitivity when sampling the cellular compartment compared with the non-cellular compartment of peripheral blood (OR 21.69; 95% CI 3.64, 229.20; p<0.0001). In addition, we show that children with autoimmunity are more likely to test positive for enterovirus RNA than those without autoimmunity (OR 11.60; 95% CI 1.89, 126.90; p=0.0065). Furthermore, we found that individuals carrying the predisposing allele (946Thr) of the common variant in IFIH1 (rs1990760, Thr946Ala) are more likely to test positive for enterovirus in peripheral blood (OR 3.07; 95% CI 1.02, 8.58; p=0.045). In contrast, using immunohistochemistry, there was no correlation between the common variant in IFIH1 and detection of enteroviral VP1 protein in the pancreatic islets of donors with type 1 diabetes. CONCLUSIONS/INTERPRETATION: Our data indicate that, in peripheral blood, antigen-presenting cells are the predominant source of enterovirus infection, and that infection is correlated with disease stage and genetic predisposition, thereby supporting a role for enterovirus infection prior to disease onset.


Asunto(s)
Diabetes Mellitus Tipo 1 , Infecciones por Enterovirus , Enterovirus , Insulinas , Adulto , Alelos , Autoanticuerpos/metabolismo , Niño , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Enterovirus/genética , Infecciones por Enterovirus/genética , Predisposición Genética a la Enfermedad , Humanos , Insulinas/genética , Insulinas/metabolismo , Helicasa Inducida por Interferón IFIH1/genética , Helicasa Inducida por Interferón IFIH1/metabolismo , Leucocitos Mononucleares/metabolismo , ARN
3.
Nat Immunol ; 21(10): 1244-1255, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32747817

RESUMEN

Follicular helper T (TFH) cells are implicated in type 1 diabetes (T1D), and their development has been linked to CD28 costimulation. We tested whether TFH cells were decreased by costimulation blockade using the CTLA-4-immunoglobulin (Ig) fusion protein (abatacept) in a mouse model of diabetes and in individuals with new-onset T1D. Unbiased bioinformatics analysis identified that inducible costimulatory molecule (ICOS)+ TFH cells and other ICOS+ populations, including peripheral helper T cells, were highly sensitive to costimulation blockade. We used pretreatment TFH profiles to derive a model that could predict clinical response to abatacept in individuals with T1D. Using two independent approaches, we demonstrated that higher frequencies of ICOS+ TFH cells at baseline were associated with a poor clinical response following abatacept administration. Therefore, TFH analysis may represent a new stratification tool, permitting the identification of individuals most likely to benefit from costimulation blockade.


Asunto(s)
Abatacept/uso terapéutico , Antígenos CD28/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Centro Germinal/inmunología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia/métodos , Linfocitos T Colaboradores-Inductores/inmunología , Abatacept/farmacología , Animales , Biomarcadores Farmacológicos , Antígenos CD28/genética , Células Cultivadas , Biología Computacional , Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/terapia , Modelos Animales de Enfermedad , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Resultado del Tratamiento
4.
Curr Opin Endocrinol Diabetes Obes ; 27(4): 240-247, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32618636

RESUMEN

PURPOSE OF REVIEW: The role of T cells specific for islet autoantigens is proven in pathogenesis of type 1 diabetes. Recently, there has been rapid expansion in the number of T-cell subsets identified, this has coincided with an increase in the repertoire of reported islet antigens mainly through the discovery of novel epitopes. A discussion of how these marry together is now warranted and timely. RECENT FINDINGS: In this review, we will discuss the autoreactivity against neo-epitopes. We then explore the growing array of T-cell subsets for both CD4 T cells, including follicular and peripheral T helper cells, and CD8 T cells, discussing evolution from naïve to exhausted phenotypes. Finally, we detail how subsets correlate with disease stage and loss of ß-cell function and are impacted by immunotherapy. SUMMARY: The expanding list of T-cell subsets may be potentially encouraging in terms of elucidating disease mechanisms and have a role as biomarkers for disease progression. Furthermore, T-cell subsets can be used in stratifying patients for clinical trials and for monitoring immunotherapy outcomes. However, the definition of subsets needs to be refined in order to ensure that there is a uniform approach in designating T-cell subset attributes that is globally applied.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Subgrupos de Linfocitos T/fisiología , Autoantígenos/inmunología , Autoantígenos/metabolismo , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD8-positivos/fisiología , Diabetes Mellitus Tipo 1/patología , Progresión de la Enfermedad , Humanos , Inmunoterapia/efectos adversos , Inmunoterapia/métodos , Islotes Pancreáticos/inmunología , Subgrupos de Linfocitos T/inmunología
5.
J Immunol ; 204(12): 3129-3138, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32404353

RESUMEN

We previously reported that costimulation blockade by abatacept limits the decline of ß-cell function and the frequency of circulating CD4+ central memory T cells (TCM) (CD45RO+CD62L+) in new-onset type 1 diabetes. In human subjects receiving placebo, we found a significant association between an increase in CD4+ TCM cells and the decline of ß-cell function. To extend and refine these findings, we examined changes in human CD4+ and CD8+ naive and memory T cell subsets at greater resolution using polychromatic flow and mass cytometry. In the placebo group, we successfully reproduced the original finding of a significant association between TCM and ß-cell function and extended this to other T cell subsets. Furthermore, we show that abatacept treatment significantly alters the frequencies of a majority of CD4+ conventional and regulatory T cell subsets; in general, Ag-naive subsets increase and Ag-experienced subsets decrease, whereas CD8+ T cell subsets are relatively resistant to drug effects, indicating a lesser reliance on CD28-mediated costimulation. Importantly, abatacept uncouples the relationship between changes in T cell subsets and ß-cell function that is a component of the natural history of the disease. Although these data suggest immunological markers for predicting change in ß-cell function in type 1 diabetes, the finding that abatacept blunts this relationship renders the biomarkers nonpredictive for this type of therapy. In sum, our findings point to a novel mechanism of action for this successful immunotherapy that may guide other disease-modifying approaches for type 1 diabetes.


Asunto(s)
Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Memoria Inmunológica/inmunología , Abatacept/farmacología , Linfocitos B/efectos de los fármacos , Biomarcadores/sangre , Antígenos CD28/inmunología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Células Cultivadas , Diabetes Mellitus Tipo 1/sangre , Humanos , Memoria Inmunológica/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología
6.
J Clin Invest ; 128(8): 3460-3474, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29851415

RESUMEN

In type 1 diabetes, cytotoxic CD8+ T cells with specificity for ß cell autoantigens are found in the pancreatic islets, where they are implicated in the destruction of insulin-secreting ß cells. In contrast, the disease relevance of ß cell-reactive CD8+ T cells that are detectable in the circulation, and their relationship to ß cell function, are not known. Here, we tracked multiple, circulating ß cell-reactive CD8+ T cell subsets and measured ß cell function longitudinally for 2 years, starting immediately after diagnosis of type 1 diabetes. We found that change in ß cell-specific effector memory CD8+ T cells expressing CD57 was positively correlated with C-peptide change in subjects below 12 years of age. Autoreactive CD57+ effector memory CD8+ T cells bore the signature of enhanced effector function (higher expression of granzyme B, killer-specific protein of 37 kDa, and CD16, and reduced expression of CD28) compared with their CD57- counterparts, and network association modeling indicated that the dynamics of ß cell-reactive CD57+ effector memory CD8+ T cell subsets were strongly linked. Thus, coordinated changes in circulating ß cell-specific CD8+ T cells within the CD57+ effector memory subset calibrate to functional insulin reserve in type 1 diabetes, providing a tool for immune monitoring and a mechanism-based target for immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Memoria Inmunológica , Células Secretoras de Insulina/inmunología , Adulto , Linfocitos T CD8-positivos/patología , Niño , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/terapia , Femenino , Humanos , Células Secretoras de Insulina/patología , Masculino
7.
Diabetes ; 67(4): 687-696, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29343547

RESUMEN

The signal peptide region of preproinsulin (PPI) contains epitopes targeted by HLA-A-restricted (HLA-A0201, A2402) cytotoxic T cells as part of the pathogenesis of ß-cell destruction in type 1 diabetes. We extended the discovery of the PPI epitope to disease-associated HLA-B*1801 and HLA-B*3906 (risk) and HLA-A*1101 and HLA-B*3801 (protective) alleles, revealing that four of six alleles present epitopes derived from the signal peptide region. During cotranslational translocation of PPI, its signal peptide is cleaved and retained within the endoplasmic reticulum (ER) membrane, implying it is processed for immune recognition outside of the canonical proteasome-directed pathway. Using in vitro translocation assays with specific inhibitors and gene knockout in PPI-expressing target cells, we show that PPI signal peptide antigen processing requires signal peptide peptidase (SPP). The intramembrane protease SPP generates cytoplasm-proximal epitopes, which are transporter associated with antigen processing (TAP), ER-luminal epitopes, which are TAP independent, each presented by different HLA class I molecules and N-terminal trimmed by ER aminopeptidase 1 for optimal presentation. In vivo, TAP expression is significantly upregulated and correlated with HLA class I hyperexpression in insulin-containing islets of patients with type 1 diabetes. Thus, PPI signal peptide epitopes are processed by SPP and loaded for HLA-guided immune recognition via pathways that are enhanced during disease pathogenesis.


Asunto(s)
Ácido Aspártico Endopeptidasas/inmunología , Diabetes Mellitus Tipo 1/inmunología , Epítopos/inmunología , Antígenos HLA-A/inmunología , Antígenos HLA-B/inmunología , Insulina/inmunología , Precursores de Proteínas/inmunología , Linfocitos T Citotóxicos/inmunología , Aminopeptidasas , Ácido Aspártico Endopeptidasas/metabolismo , Línea Celular , Retículo Endoplásmico , Técnicas de Inactivación de Genes , Predisposición Genética a la Enfermedad , Antígeno HLA-A11/inmunología , Antígeno HLA-A2/inmunología , Antígeno HLA-A24/inmunología , Humanos , Técnicas In Vitro , Insulina/metabolismo , Antígenos de Histocompatibilidad Menor , Factores Protectores , Precursores de Proteínas/metabolismo , Señales de Clasificación de Proteína , Transporte de Proteínas , Factores de Riesgo
8.
Sci Transl Med ; 9(402)2017 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-28794283

RESUMEN

Immunotherapy using short immunogenic peptides of disease-related autoantigens restores immune tolerance in preclinical disease models. We studied safety and mechanistic effects of injecting human leukocyte antigen-DR4(DRB1*0401)-restricted immunodominant proinsulin peptide intradermally every 2 or 4 weeks for 6 months in newly diagnosed type 1 diabetes patients. Treatment was well tolerated with no systemic or local hypersensitivity. Placebo subjects showed a significant decline in stimulated C-peptide (measuring insulin reserve) at 3, 6, 9, and 12 months versus baseline, whereas no significant change was seen in the 4-weekly peptide group at these time points or the 2-weekly group at 3, 6, and 9 months. The placebo group's daily insulin use increased by 50% over 12 months but remained unchanged in the intervention groups. C-peptide retention in treated subjects was associated with proinsulin-stimulated interleukin-10 production, increased FoxP3 expression by regulatory T cells, low baseline levels of activated ß cell-specific CD8 T cells, and favorable ß cell stress markers (proinsulin/C-peptide ratio). Thus, proinsulin peptide immunotherapy is safe, does not accelerate decline in ß cell function, and is associated with antigen-specific and nonspecific immune modulation.


Asunto(s)
Diabetes Mellitus Tipo 1/terapia , Inmunoterapia/métodos , Péptidos/uso terapéutico , Proinsulina/uso terapéutico , Adolescente , Adulto , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Péptido C/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Método Doble Ciego , Femenino , Humanos , Inmunofenotipificación , Masculino , Persona de Mediana Edad , Linfocitos T Reguladores/metabolismo , Adulto Joven
9.
J Autoimmun ; 84: 12-20, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28803690

RESUMEN

The mechanism by which immune tolerance is breached in autoimmune disease is poorly understood. One possibility is that post-translational modification of self-antigens leads to peripheral recognition of neo-epitopes against which central and peripheral tolerance is inadequate. Accumulating evidence points to multiple mechanisms through which non-germline encoded sequences can give rise to these non-conventional epitopes which in turn engage the immune system as T cell targets. In particular, where these modifications alter the rules of epitope engagement with MHC molecules, such non-conventional epitopes offer a persuasive explanation for associations between specific HLA alleles and autoimmune diseases. In this review article, we discuss current understanding of mechanisms through which non-conventional epitopes may be generated, focusing on several recently described pathways that can transpose germline-encoded sequences. We contextualise these discoveries around type 1 diabetes, the prototypic organ-specific autoimmune disease in which specific HLA-DQ molecules confer high risk. Non-conventional epitopes have the potential to act as tolerance breakers or disease drivers in type 1 diabetes, prompting a timely re-evaluation of models of a etiopathogenesis. Future studies are required to elucidate the disease-relevance of a range of potential non-germline epitopes and their relationship to the natural peptide repertoire.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Epítopos de Linfocito T/metabolismo , Linfocitos T/inmunología , Animales , Autoantígenos/inmunología , Autoinmunidad , Diabetes Mellitus Tipo 1/genética , Antígenos HLA-DQ/genética , Antígenos HLA-DQ/metabolismo , Humanos , Tolerancia Inmunológica , Riesgo
10.
J Autoimmun ; 66: 7-16, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26343335

RESUMEN

Type 1 diabetes was one of the earliest disorders to be associated with the phenomenon of autoimmunity and is one of the most studied organ-specific autoimmune diseases at the epidemiologic, immunologic and genetic level. Despite this, and the emergence of a plethora of strategies for trying to intervene in, or prevent the disease, it remains at some distance from being reliably and safely tractable by immunotherapy, a source of great frustration in this research field. In this article we review some of the key concepts that might impact upon this lack of success in the clinic going forward. These include new insights into autoreactive CD4 and CD8 T cell biology and a discussion of the concept of disease heterogeneity as it applies to type 1 diabetes. The onset of disease is characterised by a delicate equilibrium of proinflammatory and regulatory T cells, which we have termed "balanced autoreactive set-point", and which may be amenable to antigen-specific immunotherapies that alter the rate of disease progression. Advances in the characterization of T cells, especially at the single cell level, could be rewarding, notably from the vantage point of biomarker and surrogate discovery. A better understanding of T cell targeting, autoantigen processing and the ß-cell:immune interface is also needed, although access to diseased tissues is a major limitation in this effort. Finally, the existence of disease heterogeneity is an emerging theme in this and other complex immunopathologies, and could be both a blessing (finding the right drug for the right person) and a hindrance (compromising the power of early-stage trials of emerging therapeutics).


Asunto(s)
Autoantígenos/metabolismo , Autoinmunidad , Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Células Secretoras de Insulina/inmunología , Linfocitos T Reguladores/inmunología , Factores de Edad , Anticuerpos Monoclonales/uso terapéutico , Presentación de Antígeno , Autoantígenos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/terapia , Humanos , Factores Inmunológicos/uso terapéutico , Inmunoterapia , Inflamación/inmunología , Células Secretoras de Insulina/metabolismo , Activación de Linfocitos , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Reguladores/metabolismo
11.
J Immunol ; 195(3): 1162-70, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26085686

RESUMEN

Human monocytes comprise three distinct subsets, defined by their relative expression of CD14 and CD16. These subsets appear to have different functional roles within homeostasis and inflammation, but little is known about the manner in which they interact with macro- and microvascular endothelial cells, a key enabling component for the fulfillment of their functional roles. In the present study, we examined the locomotory behavior of the three major human monocyte subsets over human endothelial monolayers subjected to physiologically relevant levels of shear flow in vitro. Each subset was shown to preferentially perform different types of locomotory behavior in a resting state. A long-range crawling behavior, similar to the "patrolling" behavior of murine Ly6C(-) monocytes, was observed in CD14(+)CD16(-) and CD14(dim)CD16(+) monocytes, but not in CD14(+)CD16(+) monocytes. CD14(dim)CD16(+) and CD14(+)CD16(+) monocytes showed a preference for adhering to microvascular over macrovascular endothelium, whereas CD14(+)CD16(-) monocytes showed the opposite. Transendothelial migration was not observed in CD14(dim)CD16(+) monocytes during the 30-min observation period. Long-range crawling behavior in CD14(dim)CD16(+) monocytes was abrogated by blockade of ICAM1, VCAM1, or CX3CL1, in contrast with CD14(+)CD16(-) monocytes, which only required ICAM1 for this behavior. These studies indicate the existence of subtype-specific human monocyte migratory behavior patterns with distinct adhesion molecule dependence, which may assist in elucidating their physiological function and relevance to disease.


Asunto(s)
Adhesión Celular/inmunología , Movimiento Celular/inmunología , Células Endoteliales de la Vena Umbilical Humana/inmunología , Monocitos/inmunología , Animales , Antígenos Ly/genética , Células Cultivadas , Quimiocina CX3CL1/genética , Técnicas de Cocultivo , Proteínas Ligadas a GPI/biosíntesis , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Inflamación/inmunología , Molécula 1 de Adhesión Intercelular/genética , Receptores de Lipopolisacáridos/biosíntesis , Ratones , Receptores de IgG/biosíntesis , Molécula 1 de Adhesión Celular Vascular/genética
12.
Diabetes ; 64(3): 916-925, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25249579

RESUMEN

Autoreactive CD8 T cells play a central role in the destruction of pancreatic islet ß-cells that leads to type 1 diabetes, yet the key features of this immune-mediated process remain poorly defined. In this study, we combined high-definition polychromatic flow cytometry with ultrasensitive peptide-human leukocyte antigen class I tetramer staining to quantify and characterize ß-cell-specific CD8 T cell populations in patients with recent-onset type 1 diabetes and healthy control subjects. Remarkably, we found that ß-cell-specific CD8 T cell frequencies in peripheral blood were similar between subject groups. In contrast to healthy control subjects, however, patients with newly diagnosed type 1 diabetes displayed hallmarks of antigen-driven expansion uniquely within the ß-cell-specific CD8 T cell compartment. Molecular analysis of selected ß-cell-specific CD8 T cell populations further revealed highly skewed oligoclonal T cell receptor repertoires comprising exclusively private clonotypes. Collectively, these data identify novel and distinctive features of disease-relevant CD8 T cells that inform the immunopathogenesis of type 1 diabetes.


Asunto(s)
Autoantígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Células Secretoras de Insulina/inmunología , Adulto , Linfocitos T CD8-positivos/citología , Diferenciación Celular/fisiología , Femenino , Citometría de Flujo , Glutamato Descarboxilasa/inmunología , Humanos , Células Secretoras de Insulina/citología , Masculino , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/inmunología
13.
Diabetes ; 62(1): 205-13, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22936177

RESUMEN

The end-stage immunopathology of type 1 diabetes resulting in ß-cell destruction appears to be strongly dominated by cytotoxic CD8 T lymphocytes (CD8 T cells). However, the mechanism of cytotoxicity used by autoreactive CD8 T cells in the human setting remains unknown. Using type 1 diabetes patient-derived preproinsulin-specific CD8 T-cell clones recognizing either an HLA-A2 (A*0201) or HLA-A24 (A*2402)-restricted epitope (peptide of preproinsulin [PPI](15-24), ALWGPDPAAA; or PPI(3-11), LWMRLLPLL), we assessed the use of conventional mediators of cytotoxicity in the destruction of human ß-cells in vitro compared with virus-specific cytotoxic CD8 T-cell clones. We show that PPI-specific CD8 T-cell clones are mainly reliant upon cytotoxic degranulation for inducing ß-cell death. Furthermore, we find that in comparison with virus-specific CD8 T cells, there are differences in the killing potency of PPI-specific CD8 T cells that are not due to cell-intrinsic differences, but rather are mediated by differences in strength of signaling by peptide-HLA ligands. The study highlights the regulation of ß-cell killing as a potential point for therapeutic control, including the possibility of blocking autoreactive CD8 T-cell function without impacting upon general immune competence.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Degranulación de la Célula , Citotoxicidad Inmunológica , Células Secretoras de Insulina/patología , Insulina/inmunología , Precursores de Proteínas/inmunología , Receptores de Antígenos de Linfocitos T/fisiología , Línea Celular , Proteína Ligando Fas/fisiología , Humanos , Factor de Necrosis Tumoral alfa/fisiología , Receptor fas/fisiología
14.
Diabetes ; 61(7): 1752-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22522618

RESUMEN

Type 1 diabetes results from T cell-mediated ß-cell destruction. The HLA-A*24 class I gene confers significant risk of disease and early onset. We tested the hypothesis that HLA-A24 molecules on islet cells present preproinsulin (PPI) peptide epitopes to CD8 cytotoxic T cells (CTLs). Surrogate ß-cell lines secreting proinsulin and expressing HLA-A24 were generated and their peptide ligandome examined by mass spectrometry to discover naturally processed and HLA-A24-presented PPI epitopes. A novel PPI epitope was identified and used to generate HLA-A24 tetramers and examine the frequency of PPI-specific T cells in new-onset HLA-A*24(+) patients and control subjects. We identified a novel naturally processed and HLA-A24-presented PPI signal peptide epitope (PPI(3-11); LWMRLLPLL). HLA-A24 tetramer analysis reveals a significant expansion of PPI(3-11)-specific CD8 T cells in the blood of HLA-A*24(+) recent-onset patients compared with HLA-matched control subjects. Moreover, a patient-derived PPI(3-11)-specific CD8 T-cell clone shows a proinflammatory phenotype and kills surrogate ß-cells and human HLA-A*24(+) islet cells in vitro. These results indicate that the type 1 diabetes susceptibility molecule HLA-A24 presents a naturally processed PPI signal peptide epitope. PPI-specific, HLA-A24-restricted CD8 T cells are expanded in patients with recent-onset disease. Human islet cells process and present PPI(3-11), rendering themselves targets for CTL-mediated killing.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Antígeno HLA-A24/inmunología , Células Secretoras de Insulina/inmunología , Insulina/inmunología , Precursores de Proteínas/inmunología , Señales de Clasificación de Proteína , Adulto , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Muerte Celular/inmunología , Línea Celular , Epítopos de Linfocito T/inmunología , Femenino , Glutamato Descarboxilasa/inmunología , Humanos , Insulina/sangre , Masculino , Persona de Mediana Edad , Precursores de Proteínas/sangre , Adulto Joven
15.
BMC Cancer ; 11: 509, 2011 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-22152101

RESUMEN

BACKGROUND: The activity of the epidermal growth factor receptor (EGFR)-directed monoclonal antibody cetuximab combined with oxaliplatin/leucovorin/5-fluorouracil (FUFOX) was assessed in first-line metastatic gastric and oesophago-gastric junction (OGJ) cancer in a prospective phase II study showing a promising objective tumour response rate of 65% and a low mutation frequency of KRAS (3%). The aim of the correlative tumour tissue studies was to investigate the relationship between EGFR gene copy numbers, activation of the EGFR pathway, expression and mutation of E-cadherin, V600E BRAF mutation and clinical outcome of patients with gastric and OGJ cancer treated with cetuximab combined with FUFOX. METHODS: Patients included in this correlative study (n = 39) were a subset of patients from the clinical phase II study. The association between EGFR gene copy number, activation of the EGFR pathway, abundance and mutation of E-cadherin which plays an important role in these disorders, BRAF mutation and clinical outcome of patients was studied. EGFR gene copy number was assessed by FISH. Expression of the phosphorylated forms of EGFR and its downstream effectors Akt and MAPK, in addition to E-cadherin was analysed by immunohistochemistry. The frequency of mutant V600E BRAF was evaluated by allele-specific PCR and the mutation profile of the E-cadherin gene CDH1 was examined by DHPLC followed by direct sequence analysis. Correlations with overall survival (OS), time to progression (TTP) and overall response rate (ORR) were assessed. RESULTS: Our study showed a significant association between increased EGFR gene copy number (≥ 4.0) and OS in gastric and OGJ cancer, indicating the possibility that patients may be selected for treatment on a genetic basis. Furthermore, a significant correlation was shown between activated EGFR and shorter TTP and ORR, but not between activated EGFR and OS. No V600E BRAF mutations were identified. On the other hand, an interesting trend between high E-cadherin expression levels and better OS was observed and two CDH1 exon 9 missense mutations (A408V and D402H) were detected. CONCLUSION: Our finding that increased EGFR gene copy numbers, activated EGFR and the E-cadherin status are potentially interesting biomarkers needs to be confirmed in larger randomized clinical trials. TRIAL REGISTRATION: Multicentre clinical study with the European Clinical Trials Database number 2004-004024-12.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Esofágicas/tratamiento farmacológico , Unión Esofagogástrica , Neoplasias Gástricas/tratamiento farmacológico , Complejo Vitamínico B/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales Humanizados , Antígenos CD , Biomarcadores de Tumor/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Cetuximab , Receptores ErbB/genética , Receptores ErbB/metabolismo , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/secundario , Femenino , Fluorouracilo/administración & dosificación , Humanos , Inmunohistoquímica , Leucovorina/administración & dosificación , Masculino , Persona de Mediana Edad , Compuestos Organoplatinos/administración & dosificación , Oxaliplatino , Estudios Prospectivos , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/secundario , Análisis de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA