Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Gene Ther ; 26(1-2): 40-56, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30514969

RESUMEN

We report the restoration of euglycaemia in chemically induced diabetic C57BL/6 mice and spontaneously diabetic Non Obese Diabetic (NOD) mice by intravenous systemic administration of a single-stranded adeno-associated virus (ssAAV2/8) codon optimised (co) vector encoding furin cleavable human proinsulin under a liver-specific promoter. There were no immunological barriers to efficacy of insulin gene therapy in chemically induced C57BL/6 mice, which enjoyed long-lasting correction of hyperglycaemia after therapy, up to 250 days. Euglycaemia was also restored in spontaneously diabetic NOD mice, although these mice required a 7-10-fold higher dose of vector to achieve similar efficacy as the C57BL/6 mice and the immunodeficient NODscid mice. We detected CD8+ T cell reactivity to insulin and mild inflammatory infiltration in the livers of gene therapy recipient NOD mice, neither of which were observed in the treated C57BL/6 mice. Efficacy of the gene therapy in NOD mice was partially improved by targeting the immune system with anti-CD4 antibody treatment, while transfer of NOD mouse AAV2/8-reactive serum to recipients prevented successful restoration of euglycaemia in AAV2/8-HLP-hINSco-treated NODscid mice. Our data indicate that both immune cells and antibodies form a barrier to successful restoration of euglycaemia in autoimmune diabetic recipient mice with insulin gene therapy, but that this barrier can be overcome by increasing the dose of vector and by suppressing immune responses.


Asunto(s)
Dependovirus/inmunología , Diabetes Mellitus Experimental/terapia , Terapia Genética/efectos adversos , Terapia de Inmunosupresión/métodos , Insulina/inmunología , Animales , Antígenos CD4/inmunología , Dependovirus/genética , Terapia Genética/métodos , Células HEK293 , Humanos , Insulina/genética , Hígado/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Linfocitos T/inmunología
2.
Front Immunol ; 9: 1006, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29867981

RESUMEN

We present a novel and readily accessible method facilitating cellular time-resolved imaging of transplanted pancreatic islets. Grafting of islets to the mouse ear pinna allows non-invasive, in vivo longitudinal imaging of events in the islets and enables improved acquisition of experimental data and use of fewer experimental animals than is possible using invasive techniques, as the same mouse can be assessed for the presence of islet infiltrating cells before and after immune intervention. We have applied this method to investigating therapeutic protection of beta cells through the well-established use of anti-CD3 injection, and have acquired unprecedented data on the nature and rapidity of the effect on the islet infiltrating T cells. We demonstrate that infusion of anti-CD3 antibody leads to immediate effects on islet infiltrating T cells in islet grafts in the pinna of the ear, and causes them to increase their speed and displacement within 20 min of infusion. This technique overcomes several technical challenges associated with intravital imaging of pancreatic immune responses and facilitates routine study of beta islet cell development, differentiation, and function in health and disease.


Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Pabellón Auricular/inmunología , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/inmunología , Muromonab-CD3/uso terapéutico , Animales , Autoinmunidad , Modelos Animales de Enfermedad , Pabellón Auricular/diagnóstico por imagen , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Microscopía de Fluorescencia por Excitación Multifotónica , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Trasplante Isogénico
3.
Biosci Rep ; 37(4)2017 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-28798194

RESUMEN

Metabolism is of central importance for T cell survival and differentiation. It is well known that T cells cannot function in the absence of glucose, but it is less clear how they respond to excessive levels of glucose. In the present study, we investigated how increasing levels of glucose affect T-cell-mediated immune responses. We examined the effects of increased levels of glucose on CD8+ T-cell behaviour in vitro by assessing activation and cytokine production, as well as oxygen consumption rate (OCR), extracellular acidification rate (ECAR) and intracellular signalling. In addition, we assessed in vivo proliferation, cytokine production and cytolytic activity of cells in chemically induced diabetic C57BL/6 mice. Elevated levels of glucose in in vitro cultures had modest effects on proliferation and cytokine production, while in vivo hyperglycaemia had no effect on CD8+ T-cell proliferation, interferon γ (IFNγ) production or cytolytic killing.


Asunto(s)
Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Experimental/inmunología , Hiperglucemia/inmunología , Activación de Linfocitos , Animales , Linfocitos T CD8-positivos/patología , Diabetes Mellitus Experimental/patología , Hiperglucemia/patología , Interferón gamma/inmunología , Ratones , Consumo de Oxígeno/inmunología
4.
Immunology ; 151(2): 248-260, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28211040

RESUMEN

T cells play a key role in the pathogenesis of type 1 diabetes, and targeting the CD3 component of the T-cell receptor complex provides one therapeutic approach. Anti-CD3 treatment can reverse overt disease in spontaneously diabetic non-obese diabetic mice, an effect proposed to, at least in part, be caused by a selective depletion of pathogenic cells. We have used a transfer model to further investigate the effects of anti-CD3 treatment on green fluorescent protein (GFP)+ islet-specific effector T cells in vivo. The GFP expression allowed us to isolate the known effectors at different time-points during treatment to assess cell presence in various organs as well as gene expression and cytokine production. We find, in this model, that anti-CD3 treatment does not preferentially deplete the transferred effector cells, but instead inhibits their metabolic function and their production of interferon-γ. Programmed cell death protein 1 (PD-1) expression was up-regulated on the effector cells from anti-CD3-treated mice, and diabetes induced through anti-PD-L1 antibody could only be reversed with anti-CD3 antibody if the anti-CD3 treatment lasted beyond the point when the anti-PD-L1 antibody was washed out of the system. This suggests that PD-1/PD-L1 interaction plays an important role in the anti-CD3 antibody mediated protection. Our data demonstrate an additional mechanism by which anti-CD3 therapy can reverse diabetogenesis.


Asunto(s)
Anticuerpos/inmunología , Complejo CD3/inmunología , Inflamación/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Regulación hacia Arriba , Animales , Femenino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Receptor de Muerte Celular Programada 1/biosíntesis , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología
5.
J Biol Chem ; 292(15): 6281-6290, 2017 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-28188290

RESUMEN

The adaptor protein Src homology 2 domain-containing leukocyte phosphoprotein of 76 kDa (SLP-76) plays a crucial role in T cell activation by linking antigen receptor (T cell receptor, TCR) signals to downstream pathways. At its N terminus, SLP-76 has three key tyrosines (Tyr-113, Tyr-128, and Tyr-145, "3Y") as well as a sterile α motif (SAM) domain whose function is unclear. We showed previously that the SAM domain has two binding regions that mediate dimer and oligomer formation. In this study, we have identified SAM domain-carrying non-receptor tyrosine kinase, activated Cdc42-associated tyrosine kinase 1 (ACK1; also known as Tnk2, tyrosine kinase non-receptor 2) as a novel binding partner of SLP-76. Co-precipitation, laser-scanning confocal microscopy, and in situ proximity analysis confirmed the binding of ACK1 to SLP-76. Further, the interaction was induced in response to the anti-TCR ligation and abrogated by the deletion of SLP-76 SAM domain (ΔSAM) or mutation of Tyr-113, Tyr-128, and Tyr-145 to phenylalanine (3Y3F). ACK1 induced phosphorylation of the SLP-76 N-terminal tyrosines (3Y) dependent on the SAM domain. Further, ACK1 promoted calcium flux and NFAT-AP1 promoter activity and decreased the motility of murine CD4+ primary T cells on ICAM-1-coated plates, an event reversed by a small molecule inhibitor of ACK1 (AIM-100). These findings identify ACK1 as a novel SLP-76-associated protein-tyrosine kinase that modulates early activation events in T cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Activación de Linfocitos/fisiología , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Secuencias de Aminoácidos , Sustitución de Aminoácidos , Animales , Humanos , Células Jurkat , Ratones , Mutación Missense , Fosfoproteínas/genética , Fosfoproteínas/inmunología , Fosforilación/fisiología , Dominios Proteicos , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Tirosina
6.
J Gene Med ; 18(10): 312-321, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27572655

RESUMEN

BACKGROUND: The lack of an ideal cell type that can be easily acquired, modified to produce insulin, and re-implanted has been a limitation for ex vivo insulin gene therapy. Canine diabetes is currently treated with human insulin and is a good model for human diabetes. Mesenchymal stromal cells (MSCs) are a promising candidate cell type for gene therapy. In the present study, we optimised insulin production using lentiviral transduced canine MSCs (cMSCs), aiming to evaluate their ability for use as surrogate beta cells. METHODS: Canine MSCs were derived from bone marrow and validated by measuring the expression of MSC lineage specific markers. Lentivirus vectors encoding the proinsulin gene (with or without a Kozak sequence) under the control of spleen focus forming virus, cytomegalovirus, elongation factor 1α and simian virus 40 promotors were generated and used to transduce primary cMSCs and a hepatocyte cell line. The insulin-producing capacity of transduced primary cMSCs was assessed by measuring the concentration of C-peptide produced. RESULTS: Primary cMSC could be readily expanded in culture and efficiently transduced using lentiviral vectors encoding proinsulin. Increasing the multiplicity of infection from 3 to 20 led to an increase in C-peptide secretion (from 1700 to 4000 pmol/l). The spleen focus forming virus promoter conferred the strongest transcriptional ability. CONCLUSIONS: The results of the present study suggest that optimised lentiviral transduction of the insulin gene into primary cMSCs renders these cells capable of secreting insulin over both the short- and long-term, in sufficient quantities in vitro to support their potential use in insulin gene therapy.


Asunto(s)
Expresión Génica , Insulina/genética , Lentivirus/genética , Células Madre Mesenquimatosas/metabolismo , Regiones Promotoras Genéticas/genética , Animales , Células de la Médula Ósea/metabolismo , Línea Celular Tumoral , Células Cultivadas , Perros , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/genética , Células HEK293 , Hepatocitos/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Proinsulina/genética , Proinsulina/metabolismo
7.
PLoS One ; 11(1): e0146516, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26783747

RESUMEN

Type 1 diabetes is caused by the destruction of insulin producing beta cells by the immune system. The p110δ isoform of PI3K is expressed primarily in cells of haematopoietic origin and the catalytic activity of p110δ is important for the activation of these cells. Targeting of this pathway offers an opportunity to reduce immune cell activity without unwanted side effects. We have explored the effects of a specific p110δ isoform inhibitor, IC87114, on diabetogenic T cells both in vitro and in vivo, and find that although pharmacological inhibition of p110δ has a considerable impact on the production of pro-inflammatory cytokines, it does not delay the onset of diabetes after adoptive transfer of diabetogenic cells. Further, we demonstrate that combination treatment with CTLA4-Ig does not improve the efficacy of treatment, but instead attenuates the protective effects seen with CTLA4-Ig treatment alone. Our results suggest that decreased IL-10 production by Foxp3+ CD4+ T cells in the presence of IC87114 negates individual anti-inflammatory effects of IC8114 and CTLA4-Ig.


Asunto(s)
Adenina/análogos & derivados , Diferenciación Celular/efectos de los fármacos , Citocinas/biosíntesis , Diabetes Mellitus Tipo 1/inmunología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Quinazolinas/farmacología , Linfocitos T/efectos de los fármacos , Adenina/farmacología , Animales , Diferenciación Celular/inmunología , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase I , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Linfocitos T/fisiología
8.
Mol Cell ; 59(5): 840-9, 2015 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-26321253

RESUMEN

While immune cell adaptors regulate proximal T cell signaling, direct regulation of the nuclear pore complex (NPC) has not been reported. NPC has cytoplasmic filaments composed of RanGAP1 and RanBP2 with the potential to interact with cytoplasmic mediators. Here, we show that the immune cell adaptor SLP-76 binds directly to SUMO-RanGAP1 of cytoplasmic fibrils of the NPC, and that this interaction is needed for optimal NFATc1 and NF-κB p65 nuclear entry in T cells. Transmission electron microscopy showed anti-SLP-76 cytoplasmic labeling of the majority of NPCs in anti-CD3 activated T cells. Further, SUMO-RanGAP1 bound to the N-terminal lysine 56 of SLP-76 where the interaction was needed for optimal RanGAP1-NPC localization and GAP exchange activity. While the SLP-76-RanGAP1 (K56E) mutant had no effect on proximal signaling, it impaired NF-ATc1 and p65/RelA nuclear entry and in vivo responses to OVA peptide. Overall, we have identified SLP-76 as a direct regulator of nuclear pore function in T cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Fosfoproteínas/inmunología , Fosfoproteínas/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Transporte Activo de Núcleo Celular , Animales , Línea Celular , Humanos , Células Jurkat , Ratones , Microscopía Electrónica de Transmisión , Factores de Transcripción NFATC/metabolismo , Poro Nuclear/metabolismo , Unión Proteica , Linfocitos T/ultraestructura , Factor de Transcripción ReIA/metabolismo
9.
Biochem J ; 430(2): 207-13, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20629633

RESUMEN

RASSF7, a member of the N-terminal Ras association domain family, has increased expression in various cancers and, on the basis of our previous work in Xenopus embryos, may be a regulator of mitosis. In the present study, we address, for the first time, the role of human RASSF7 in mitosis. We demonstrate that RASSF7 is expressed in a broad range of different cell types and that this expression could be enhanced following exposure to hypoxia. Knocking down RASSF7 in human cell lines inhibited cell growth and induced defects in mitosis, including aberrant spindle formation and a failure in chromosomal congression. In order to understand the molecular basis of the defects in more detail, we analysed the activity of mitotic signalling proteins and found that activation of Aurora B did not occur in cells in which RASSF7 was knocked down. We also show that endogenous RASSF7 protein localizes to the centrosome and demonstrate using microtubule-regrowth assays that RASSF7 is an important regulator of microtubule dynamics. On the basis of these observations, we propose that, owing to its key role in regulating the microtubule cytoskeleton, RASSF7 is required for mitosis in human cells.


Asunto(s)
Segregación Cromosómica , Microtúbulos/metabolismo , Mitosis , Proteínas Serina-Treonina Quinasas/metabolismo , Huso Acromático/metabolismo , Factores de Transcripción/metabolismo , Animales , Aurora Quinasa B , Aurora Quinasas , Línea Celular , Centrosoma/metabolismo , Células HeLa , Humanos , Ratones , Microtúbulos/genética , Proteínas Serina-Treonina Quinasas/genética , Transporte de Proteínas , Huso Acromático/genética , Factores de Transcripción/genética
10.
Biochem J ; 425(2): 303-11, 2009 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-20025613

RESUMEN

The RASSF (Ras-association domain family) has recently gained several new members and now contains ten proteins (RASSF1-10), several of which are potential tumour suppressors. The family can be split into two groups, the classical RASSF proteins (RASSF1-6) and the four recently added N-terminal RASSF proteins (RASSF7-10). The N-terminal RASSF proteins have a number of differences from the classical RASSF members and represent a newly defined set of potential Ras effectors. They have been linked to key biological processes, including cell death, proliferation, microtubule stability, promoter methylation, vesicle trafficking and response to hypoxia. Two members of the N-terminal RASSF family have also been highlighted as potential tumour suppressors. The present review will summarize what is known about the N-terminal RASSF proteins, addressing their function and possible links to cancer formation. It will also compare the N-terminal RASSF proteins with the classical RASSF proteins and ask whether the N-terminal RASSF proteins should be considered as genuine members or imposters in the RASSF family.


Asunto(s)
Factores de Transcripción/fisiología , Proteínas Supresoras de Tumor/fisiología , Proteínas de Transporte Vesicular/fisiología , Fenómenos Fisiológicos Celulares , Humanos , Neoplasias/etiología , Estructura Terciaria de Proteína , Factores de Transcripción/química , Factores de Transcripción/clasificación , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/clasificación , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/clasificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...