Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 7: 44661, 2017 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-28300170

RESUMEN

CD8+ T cells directed against beta cell autoantigens are considered relevant for the pathogenesis of type 1 diabetes. Using single cell T cell receptor sequencing of CD8+ T cells specific for the IGRP265-273 epitope, we examined whether there was expansion of clonotypes and sharing of T cell receptor chains in autoreactive CD8+ T cell repertoires. HLA-A*0201 positive type 1 diabetes patients (n = 19) and controls (n = 18) were analysed. TCR α- and ß-chain sequences of 418 patient-derived IGRP265-273-multimer+ CD8+ T cells representing 48 clonotypes were obtained. Expanded populations of IGRP265-273-specific CD8+ T cells with dominant clonotypes that had TCR α-chains shared across patients were observed. The SGGSNYKLTF motif corresponding to TRAJ53 was contained in 384 (91.9%) cells, and in 20 (41.7%) patient-derived clonotypes. TRAJ53 together with TRAV29/DV5 was found in 15 (31.3%) clonotypes. Using next generation TCR α-chain sequencing, we found enrichment of one of these TCR α-chains in the memory CD8+ T cells of patients as compared to healthy controls. CD8+ T cell clones bearing the enriched motifs mediated antigen-specific target cell lysis. We provide the first evidence for restriction of T cell receptor motifs in the alpha chain of human CD8+ T cells with specificity to a beta cell antigen.


Asunto(s)
Autoantígenos/metabolismo , Linfocitos T CD8-positivos/inmunología , Glucosa-6-Fosfatasa/metabolismo , Islotes Pancreáticos/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Células Clonales , Femenino , Glucosa-6-Fosfatasa/química , Humanos , Masculino , Análisis de Secuencia de Proteína , Adulto Joven
2.
Sci Transl Med ; 9(378)2017 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-28228602

RESUMEN

Autoimmune diabetes is marked by sensitization to ß cell self-antigens in childhood. We longitudinally followed at-risk children from infancy and performed single-cell gene expression in ß cell antigen-responsive CD4+ T cells through pre- and established autoimmune phases. A striking divergence in the gene signature of ß cell antigen-responsive naïve CD4+ T cells from children who developed ß cell autoimmunity was found in infancy, well before the appearance of ß cell antigen-specific memory T cells or autoantibodies. The signature resembled a pre-T helper 1 (TH1)/TH17/T follicular helper cell response with expression of CCR6, IL21, TBX21, TNF, RORC, EGR2, TGFB1, and ICOS, in the absence of FOXP3, IL17, and other cytokines. The cells transitioned to an IFNG-TH1 memory phenotype with the emergence of autoantibodies. We suggest that the divergent naïve T cell response is a consequence of genetic or environmental priming during unfavorable perinatal exposures and that the signature will guide future efforts to detect and prevent ß cell autoimmunity.


Asunto(s)
Autoantígenos/inmunología , Autoinmunidad , Linfocitos T CD4-Positivos/inmunología , Células Secretoras de Insulina/inmunología , Autoanticuerpos/biosíntesis , Autoinmunidad/efectos de los fármacos , Autoinmunidad/genética , Linfocitos T CD4-Positivos/efectos de los fármacos , Perfilación de la Expresión Génica , Glutamato Descarboxilasa/metabolismo , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Lactante , Células Secretoras de Insulina/efectos de los fármacos , Toxina Tetánica/farmacología , Células TH1/inmunología , Transcripción Genética/efectos de los fármacos
3.
Acta Biomater ; 44: 178-87, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27506126

RESUMEN

UNLABELLED: Intrahepatic transplantation of allogeneic pancreatic islets offers a promising therapy for type 1 diabetes. However, long-term insulin independency is often not achieved due to severe islet loss shortly after transplantation. To improve islet survival and function, extrahepatic biomaterial-assisted transplantation of pancreatic islets to alternative sites has been suggested. Herein, we present macroporous, star-shaped poly(ethylene glycol) (starPEG)-heparin cryogel scaffolds, covalently modified with adhesion peptides, for the housing of pancreatic islets in three-dimensional (3D) co-culture with adherent mesenchymal stromal cells (MSC) as accessory cells. The implantable biohybrid scaffolds provide efficient transport properties, mechanical protection, and a supportive extracellular environment as a desirable niche for the islets. MSC colonized the cryogel scaffolds and produced extracellular matrix proteins that are important components of the natural islet microenvironment known to facilitate matrix-cell interactions and to prevent cellular stress. Islets survived the seeding procedure into the cryogel scaffolds and secreted insulin after glucose stimulation in vitro. In a rodent model, intact islets and MSC could be visualized within the scaffolds seven days after subcutaneous transplantation. Overall, this demonstrates the potential of customized macroporous starPEG-heparin cryogel scaffolds in combination with MSC to serve as a multifunctional islet supportive carrier for transplantation applications. STATEMENT OF SIGNIFICANCE: Diabetes results in the insufficient production of insulin by the pancreatic ß-cells in the islets of Langerhans. Transplantation of pancreatic islets offers valuable options for treating the disease; however, many transplanted islets often do not survive the transplantation or die shortly thereafter. Co-transplanted, supporting cells and biomaterials can be instrumental for improving islet survival, function and protection from the immune system. In the present study, islet supportive hydrogel sponges were explored for the co-transplantation of islets and mesenchymal stromal cells. Survival and continued function of the supported islets were demonstrated in vitro. The in vivo feasibility of the approach was shown by transplantation in a mouse model.


Asunto(s)
Materiales Biocompatibles/farmacología , Criogeles/farmacología , Islotes Pancreáticos/citología , Células Madre Mesenquimatosas/citología , Animales , Supervivencia Celular/efectos de los fármacos , Heparina/química , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Trasplante de Islotes Pancreáticos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones Endogámicos C57BL , Polietilenglicoles/química , Porosidad , Sus scrofa , Ingeniería de Tejidos , Andamios del Tejido/química , Trasplante Isogénico
4.
Clin Immunol ; 157(2): 216-25, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25728493

RESUMEN

Quantification of antigen-specific CD8(+) T cells is important for monitoring infection, vaccination, and response to therapy in cancer and immune-mediated diseases. Cytokine enzyme-linked-immunospot (ELISpot) assays are often used for this purpose. We found that substantial spot formation in IFNγ ELISpot assays occurred independently of CD8(+) T cells even when classical MHC class I restricted peptides are used for stimulation. Using fractionated cells and intracellular cytokine staining, the non-CD8(+) T cell IFNγ production was attributed to the CD4(+) T cell fraction. We therefore refined a cell line-based ELISpot assay combining HLA-A*0201 expressing K562 cells for antigen presentation with purified CD8(+) T cells and demonstrated that it specifically detected CD8(+) T cell responses with detection limits comparable to traditional ELISpot assays and dextramer-based quantification. The assay was further adapted to whole antigen responses with antigen (pre-proinsulin)-expressing HLA-A*0201K562 cells. Thus, we revealed and corrected a weak spot of the CD8(+) ELISpot assay.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Ensayo de Immunospot Ligado a Enzimas/métodos , Antígeno HLA-A2/inmunología , Adolescente , Animales , Estudios de Casos y Controles , Niño , Femenino , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Interferón gamma/inmunología , Células K562 , Masculino , Ratones , Ratones Endogámicos NOD , Adulto Joven
5.
Cytotherapy ; 17(4): 473-86, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25573333

RESUMEN

BACKGROUND AIMS: Mouse models indicate that adoptive transfer of regulatory T cells (Treg) may suppress graft-versus-host-disease (GvHD) while preserving graft-versus-leukemia reactions. We aimed to develop a protocol for the efficient isolation and in vitro expansion of donor-derived Treg and to establish the proof-of-concept for the clinical application of ex vivo-generated Treg preparations in five patients with otherwise treatment-refractory chronic GvHD (cGvHD). METHODS: Allogeneic Treg were isolated from unstimulated leukapheresis products of the corresponding human leukocyte antigen-matched donors by use of clinical-grade magnetic-activated bead sorting. To increase the amount and purity, Treg were cultivated for 7-12 days and infused after a median time of 35 months after allogeneic hematopoietic cell transplantation. RESULTS: Final products contained Treg with a median purity of 84.1% CD4(+)CD25(high)CD127(low)FOXP3(+)of CD45(+) cells and a mean quantity of 2.4 × 10(6) Treg per kg body wt. All isolated cell products showed in vitro suppressive activity. On transfusion, two of five patients showed a clinical response with improvement of cGvHD symptoms. The other three patients showed stable cGvHD symptoms for up to 21 months. In four of five patients, increased counts of Treg were detectable on Treg transfusion, immunosuppressive treatment could be reduced and suppression of CD69 activation marker expression on T-effector cells was observed. However, one patient had development of malignant melanoma and another patient had Bowen skin cancer 4 months and 11 months after Treg transfusion, respectively. CONCLUSIONS: We demonstrate a feasible and reproducible approach of isolating functional Treg in high quantity and purity for clinical application and show opportunities and risks of adoptive Treg transfer into patients with cGvHD.


Asunto(s)
Traslado Adoptivo/métodos , Enfermedad Injerto contra Huésped/terapia , Terapia de Inmunosupresión/métodos , Linfocitos T Reguladores/trasplante , Adulto , Animales , Células Cultivadas , Femenino , Trasplante de Células Madre Hematopoyéticas , Humanos , Leucaféresis/métodos , Masculino , Ratones , Persona de Mediana Edad , Linfocitos T Reguladores/inmunología , Donantes de Tejidos , Trasplante Homólogo
6.
Diabetologia ; 57(3): 522-31, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24253203

RESUMEN

AIMS/HYPOTHESIS: Islet transplantation is used therapeutically in a minority of patients with type 1 diabetes. Successful outcomes are hampered by early islet beta cell loss. The adjuvant co-transplantation of mesenchymal stromal cells (MSCs) has the promise to improve islet transplant outcome. METHODS: We used a syngeneic marginal islet mass transplantation model in a mouse model of diabetes. Mice received islets or islets plus 250,000 MSCs. Kidney subcapsule, intra-hepatic and intra-ocular islet transplantation sites were used. Apoptosis, vascularisation, beta cell proliferation, MSC differentiation and laminin levels were determined by immunohistochemical analysis and image quantification post-transplant. RESULTS: Glucose homeostasis after the transplantation of syngeneic islets was improved by the co-transplantation of MSCs together with islets under the kidney capsule (p = 0.01) and by intravenous infusion of MSCs after intra-hepatic islet transplantation (p = 0.05). MSC co-transplantation resulted in reduced islet apoptosis, with reduced numbers of islet cells positive for cleaved caspase 3 being observed 14 days post-transplant. In kidney subcapsule, but not in intra-ocular islet transplant models, we observed increased re-vascularisation rates, but not increased blood vessel density in and around islets co-transplanted with MSCs compared with islets that were transplanted alone. Co-transplantation of MSCs did not increase beta cell proliferation, extracellular matrix protein laminin production or alpha cell numbers, and there was negligible MSC transdifferentiation into beta cells. CONCLUSIONS/INTERPRETATION: Co-transplantation of MSCs may lead to improved islet function and survival in the early post-transplantation period in humans receiving islet transplantation.


Asunto(s)
Diabetes Mellitus Experimental/patología , Insulina/metabolismo , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Animales , Glucemia , Proliferación Celular , Técnicas de Cocultivo , Diabetes Mellitus Experimental/inmunología , Secreción de Insulina , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica , Trasplante Isogénico
7.
J Immunol Methods ; 400-401: 13-22, 2013 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-24239865

RESUMEN

T cells have diversity in TCR, epitope recognition, and cytokine production, and can be used for immune monitoring. Furthermore, clonal expansion of TCR families in disease may provide opportunities for TCR-directed therapies. We developed methodology for sequencing expressed genes of TCR alpha and beta chains from single cells and applied this to vaccine (tetanus-toxoid)-responsive CD4(+) T cells. TCR alpha and beta chains were both successfully sequenced in 1309 (43%) of 3038 CD4(+) T cells yielding 677 different receptors. TRAV and TRBV gene usage differed between tetanus-toxoid-responsive and non-responsive cells (p=0.004 and 0.0002), and there was extensive TCR diversity in tetanus-toxoid-responsive cells within individuals. Identical TCRs could be recovered in different samples from the same subject: TCRs identified after booster vaccination were frequent in pre-booster memory T cells (31% of pre-booster TCR), and also identified in pre-booster vaccination naïve cells (6.5%). No TCR was shared between subjects, but tetanus toxoid-responsive cells sharing one of their TCR chains were observed within and between subjects. Coupling single-cell gene expression profiling to TCR sequencing revealed examples of distinct cytokine profiles in cells bearing identical TCR. Novel molecular methodology demonstrates extensive diversity of Ag-responsive CD4(+) T cells within and between individuals.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Perfilación de la Expresión Génica/métodos , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Análisis de Secuencia de ADN/métodos , Análisis de la Célula Individual/métodos , Subgrupos de Linfocitos T/inmunología , Toxoide Tetánico/inmunología , Adulto , ADN/genética , Femenino , Humanos , Memoria Inmunológica , Lactante , Masculino , Persona de Mediana Edad
8.
Diabetes ; 62(6): 2059-66, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23349478

RESUMEN

Islet autoimmunity precedes type 1 diabetes onset. We previously found that islet autoimmunity rarely starts before 6 months of age but reaches its highest incidence already at ∼1 year of age. We now examine whether homeostatic expansion and immune competence changes seen in a maturating immune system may account for this marked variation in islet autoimmunity risk in the first year of life. We found naïve proinsulin- and GAD65-responsive T cells in cord blood (CB) of healthy newborns, with highest responses observed in children with type 1 diabetes-susceptible HLA-DRB1/DQB1 genotypes. Homeostatic expansion characteristics with increased IL-7 concentrations and enhanced T-cell responsiveness to IL-7 were observed throughout the first year of life. However, the ability of antigen-presenting cells to activate naïve T cells was compromised at birth, and CB monocytes had low surface expression of CD40 and HLA class II. In contrast, antigen presentation and expression of these molecules had reached competent adult levels by the high incidence age of 8 months. We propose that temporal changes in islet autoimmunity seroconversion in infants are a consequence of the changing balance between homeostatic drive and antigen presentation competence. These findings are relevant for early prevention of type 1 diabetes.


Asunto(s)
Presentación de Antígeno/fisiología , Linfocitos T/metabolismo , Presentación de Antígeno/genética , Antígenos CD4/metabolismo , Antígenos CD40/genética , Proliferación Celular , Células Cultivadas , Citometría de Flujo , Genotipo , Antígenos HLA/genética , Cadenas beta de HLA-DQ/genética , Cadenas HLA-DRB1/genética , Haplotipos/genética , Homeostasis/genética , Homeostasis/fisiología , Humanos , Lactante , Recién Nacido , Subunidad alfa del Receptor de Interleucina-2/metabolismo
9.
J Immunol ; 189(12): 5649-58, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23129754

RESUMEN

CD4(+)CD25(+)FOXP3(+) regulatory T cells (Tregs) control the activation and expansion of alloreactive and autoreactive T cell clones. Because uncontrolled activation and expansion of autoreactive T cells occur in an IL-7-rich environment, we explored the possibility that IL-7 may affect the function of Treg. We show that the functional high-affinity IL-7R is expressed on both naive and memory Tregs, and exposure to IL-7 results in STAT-5 phosphorylation. Naive, but not memory, Tregs proliferated greatly and acquired a memory phenotype in the setting of a suppression assay when IL-7 was present. Importantly, the presence of IL-7 abrogated the capacity of Tregs to suppress proliferation of conventional T cells in response to TCR activators, including alloantigens and autoantigens. Removal of IL-7 restored the suppressive function of Tregs. Preblocking of the IL-7R on the Tregs also restored suppressor function, indicating that IL-7 directly affected Treg function. Thus, prolonged periods of homeostatic expansion can temporarily release natural regulatory brakes on T cells, thereby providing an additional mechanism for activating and expanding alloreactive and autoreactive T cells.


Asunto(s)
Autoantígenos/biosíntesis , Antígenos CD4/biosíntesis , Diferenciación Celular/inmunología , Factores de Transcripción Forkhead/biosíntesis , Interleucina-7/fisiología , Isoantígenos/biosíntesis , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Proliferación Celular , Células Cultivadas , Contraindicaciones , Humanos , Inmunofenotipificación , Interleucina-10/biosíntesis , Subunidad alfa del Receptor de Interleucina-2/biosíntesis , Activación de Linfocitos/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...