Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
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.
J Pathol ; 244(3): 283-295, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29205362

RESUMEN

The oncostatin M (OSM) receptor (OSMR) shows frequent gene copy number gains and overexpression in cervical squamous cell carcinomas (SCCs), associated with adverse clinical outcomes. In SCC cells that overexpress OSMR, the major ligand OSM induces multiple pro-malignant effects, including invasion, secretion of angiogenic factors, and metastasis. Here, we demonstrate, for the first time, that OSMR overexpression in SCC cells activates cell-autonomous feed-forward signalling, via further expression of OSMR and OSM and sustained STAT3 activation, despite expression of the negative regulator suppressor of cytokine signalling 3 (SOCS3). The pro-malignant effects associated with OSMR overexpression are critically mediated by JAK-STAT3 activation, which is induced by exogenous OSM and also by autocrine OSM-OSMR interactions. Importantly, specific inhibition of OSM-OSMR interactions by neutralizing antibodies significantly inhibits STAT3 activation and feed-forward signalling, leading to reduced invasion, angiogenesis, and metastasis. Our findings are supported by data from 1254 clinical SCC samples, in which OSMR levels correlated with multiple cognate genes, including OSM, STAT3, and downstream targets. These data strongly support the development of OSM-OSMR-blocking antibodies as biologically targeted therapies against SCCs of the cervix and other anatomical sites. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Subunidad beta del Receptor de Oncostatina M/antagonistas & inhibidores , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Neoplasias del Cuello Uterino/tratamiento farmacológico , Animales , Comunicación Autocrina , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones Endogámicos NOD , Ratones SCID , Oncostatina M/genética , Oncostatina M/metabolismo , Subunidad beta del Receptor de Oncostatina M/genética , Subunidad beta del Receptor de Oncostatina M/inmunología , Subunidad beta del Receptor de Oncostatina M/metabolismo , Fosforilación , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Regulación hacia Arriba , Neoplasias del Cuello Uterino/inmunología , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
PLoS One ; 12(8): e0181964, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28771521

RESUMEN

Microbes, including viruses, influence type 1 diabetes (T1D) development, but many such influences remain undefined. Previous work on underlying immune mechanisms has focussed on cytokines and T cells. Here, we compared two nonobese diabetic (NOD) mouse colonies, NODlow and NODhigh, differing markedly in their cumulative T1D incidence (22% vs. 90% by 30 weeks in females). NODhigh mice harbored more complex intestinal microbiota, including several pathobionts; both colonies harbored segmented filamentous bacteria (SFB), thought to suppress T1D. Young NODhigh females had increased B-cell activation in their mesenteric lymph nodes. These phenotypes were transmissible. Co-housing of NODlow with NODhigh mice after weaning did not change T1D development, but T1D incidence was increased in female offspring of co-housed NODlow mice, which were exposed to the NODhigh environment both before and after weaning. These offspring also acquired microbiota and B-cell activation approaching those of NODhigh mice. In NODlow females, the low rate of T1D was unaffected by cyclophosphamide but increased by PD-L1 blockade. Thus, environmental exposures that are innocuous later in life may promote T1D progression if acquired early during immune development, possibly by altering B-cell activation and/or PD-L1 function. Moreover, T1D suppression in NOD mice by SFB may depend on the presence of other microbial influences. The complexity of microbial immune regulation revealed in this murine model may also be relevant to the environmental regulation of human T1D.


Asunto(s)
Linfocitos B/inmunología , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 1/inmunología , Ambiente , Linfocitos T/inmunología , Animales , Linfocitos B/patología , Antígeno B7-H1/metabolismo , Citocinas/metabolismo , Diabetes Mellitus Experimental/epidemiología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/epidemiología , Diabetes Mellitus Tipo 1/patología , Femenino , Incidencia , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos NOD , Linfocitos T/patología
5.
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
6.
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
7.
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
8.
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
9.
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
10.
Trends Immunol ; 34(12): 583-91, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24054837

RESUMEN

There are three prerequisites for development of the autoimmune disease type 1 diabetes (T1D). First, ß cell-reactive T cells need to be activated; second, the response needs to be proinflammatory; and finally, immune regulation of autoreactive responses must fail. Here, we describe our current understanding of the cell types and immune mechanisms involved in each of these steps leading to T1D. Novel findings regarding ß cell involvement in its own destruction, the importance of the microbiota for instruction of the immune system, and recent data from studies in T1D patients are discussed. In addition, we summarise therapeutic approaches to T1D, and how these relate to the immune mechanisms involved in disease development.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Diabetes Mellitus Tipo 1/inmunología , Sistema Inmunológico/inmunología , Animales , Humanos , Células Secretoras de Insulina/inmunología
11.
Diabetes ; 62(9): 3132-42, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23715623

RESUMEN

In type 1 diabetes, the insulin-producing ß-cells are destroyed by the immune system. One way of restoring glucose control is to transplant ß-cells from a donor. Although this procedure may restore endogenous insulin production, immunosuppressive treatment is needed to prevent the recipient from rejecting the donor-derived islets. We investigated the possibilities of transient expression of the immunosuppressive cytokine transforming growth factor (TGF)-ß within islets to achieve long-term graft tolerance. We found that brief expression of TGF-ß prevented rejection of syngeneic islets, that there was reduction of dendritic cell (DC) activation in the graft, and that there was reduced reactivation of T cells in the graft-draining lymph nodes. In vitro exposure of bone marrow-derived DCs to TGF-ß reduced expression of costimulatory molecules CD80 and CD86, as well as production of proinflammatory cytokines such as interleukin-12 p70 in DCs, but did not alter levels of major histocompatibility complex classes I and II. Furthermore, the capacity of TGF-ß-treated bone marrow-derived DCs to activate both CD4(+) and CD8(+) T cells was reduced. Adding TGF-ß-conditioned tolerogenic DCs to the grafted islets led to long-term survival of the graft, demonstrating that TGF-ß-induced tolerogenic DCs can provide an effective means to restore immune tolerance in an already established autoimmune disease.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Trasplante de Islotes Pancreáticos/métodos , Factor de Crecimiento Transformador beta/farmacología , Animales , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular/efectos de los fármacos , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Interleucina-12/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa
12.
J Immunol ; 186(4): 2543-51, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21217013

RESUMEN

Effective therapies that prevent chronic inflammation from developing into type 1 diabetes remain elusive. In this study, we show that expression of TGF-ß for just 1 wk in inflamed islets of NOD mice significantly delays diabetes development. Time course studies demonstrated that the brief TGF-ß pulse protects only if administered when extensive ß cell destruction has occurred. Surprisingly, TGF-ß-mediated protection is not linked to enhanced Foxp3(+) regulatory T cell activity or to decreased intrapancreatic presentation of islet Ags. Instead, TGF-ß disables the transition of primed autoreactive CD8(+) T cells to cytotoxic effectors and decreases generation, or maintenance, of CD8(+) memory T cells within the pancreas, significantly impairing their diabetogenic capacity.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Epítopos de Linfocito T/inmunología , Factores de Transcripción Forkhead/biosíntesis , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/patología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología , Factor de Crecimiento Transformador beta/administración & dosificación , Animales , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Pruebas Inmunológicas de Citotoxicidad/métodos , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/prevención & control , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Factores de Transcripción Forkhead/fisiología , Memoria Inmunológica/genética , Mediadores de Inflamación/administración & dosificación , Mediadores de Inflamación/fisiología , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/patología , Factor de Crecimiento Transformador beta/fisiología
13.
Inflamm Allergy Drug Targets ; 8(2): 130-8, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19530995

RESUMEN

Incidence of autoimmune diseases is rising rapidly in the developed world and treatment of such diseases will be a major burden on Government health resources of the future. Whether systemic or organ-specific, immune cell destruction of the target tissue normally requires co-operative interaction of a many distinct immune cells. Detailed knowledge of the cells and signal pathways involved in tissue destruction is paramount to the design of novel therapeutics. Several organ-specific autoimmune diseases e.g. multiple sclerosis, rheumatoid arthritis and type 1 diabetes have long been attributed to T cell-mediated destruction of the target tissue. However, recent reports from both murine models and man have suggested that B cells are principal players in these T cell-mediated diseases. In this review, we discuss the evidence that supports a link between B cells and the autoaggressive T cell response in type 1 diabetes and how accumulating evidence suggests targeting B cells may offer a novel therapeutic strategy for this autoimmune disease.


Asunto(s)
Autoantígenos/metabolismo , Linfocitos B/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Inmunoterapia , Islotes Pancreáticos/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Anticuerpos Monoclonales de Origen Murino , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Linfocitos B/inmunología , Linfocitos B/patología , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/terapia , Humanos , Tolerancia Inmunológica , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos NOD , Rituximab , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología
14.
Diabetes ; 57(4): 909-17, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18184927

RESUMEN

OBJECTIVE: To determine the role of B-cells in promoting CD8(+) T-cell-mediated beta cell destruction in chronically inflamed islets. RESEARCH DESIGN AND METHODS-RIP: TNFalpha-NOD mice were crossed to B-cell-deficient NOD mice, and diabetes development was monitored. We used in vitro antigen presentation assays and in vivo administration of bromodeoxyuridine coupled to flow cytometry assays to assess intra-islet T-cell activation in the absence or presence of B-cells. CD4(+)Foxp3(+) activity in the absence or presence of B-cells was tested using in vivo depletion techniques. Cytokine production and apoptosis assays determined the capacity of CD8(+) T-cells transform to cytotoxic T-lymphocytes (CTLs) and survive within inflamed islets in the absence or presence of B-cells. RESULTS: B-cell deficiency significantly delayed diabetes development in chronically inflamed islets. Reintroduction of B-cells incapable of secreting immunoglobulin restored diabetes development. Both CD4(+) and CD8(+) T-cell activation was unimpaired by B-cell deficiency, and delayed disease was not due to CD4(+)Foxp3(+) T-cell suppression of T-cell responses. Instead, at the CTL transition stage, B-cell deficiency resulted in apoptosis of intra-islet CTLs. CONCLUSIONS: In inflamed islets, B-cells are central for the efficient intra-islet survival of CTLs, thereby promoting type 1 diabetes development.


Asunto(s)
Linfocitos T CD8-positivos/patología , Diabetes Mellitus Tipo 1/inmunología , Células Secretoras de Insulina/fisiología , Animales , Células Presentadoras de Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/citología , Supervivencia Celular , Citocinas/sangre , Inflamación , Células Secretoras de Insulina/patología , Depleción Linfocítica , Ratones , Ratones Endogámicos NOD
15.
Eur J Immunol ; 37(7): 1986-95, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17523133

RESUMEN

Self proteins may become autoantigenic through structural modification. We studied malondialdehydation of recombinant rat (rr) myelin oligodendrocyte glycoprotein (MOG), an autoantigen in multiple sclerosis. Malondialdehyde (MDA) modification changed protein weight and charge, the location of these adducts being mapped by Fourier transform ion cyclotron resonance. Molecular modelling revealed significant differences in the MDA-rrMOG three-dimensional structure. DBA/1 mice immunised with MDA-rrMOG developed greater proliferative responses and more severe experimental autoimmune encephalomyelitis than mice immunised with unmodified rrMOG. MDA-rrMOG was taken up more effectively by antigen-presenting cells (APC), at least partially through scavenger receptors. Exposure to MDA-rrMOG led to increased expression of IL-23, IL-12 and IL-12R, indicating a role not only for increased antigen uptake but also for activation of APC. We thus provide biochemical, structural, immunological and clinical data that suggest that the post-translationally modified form of this myelin autoantigen is a more relevant form of the molecule.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Malondialdehído/metabolismo , Glicoproteína Asociada a Mielina/inmunología , Glicoproteína Asociada a Mielina/metabolismo , Procesamiento Proteico-Postraduccional/inmunología , Animales , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Autoantígenos/química , Autoantígenos/inmunología , Autoantígenos/metabolismo , Electroforesis en Gel de Poliacrilamida , Femenino , Análisis de Fourier , Focalización Isoeléctrica , Malondialdehído/inmunología , Ratones , Proteínas de la Mielina , Glicoproteína Asociada a Mielina/química , Glicoproteína Mielina-Oligodendrócito , Reacción en Cadena de la Polimerasa , Estructura Terciaria de Proteína , Ratas , Espectrometría de Masa por Ionización de Electrospray
16.
J Immunol ; 177(8): 5105-14, 2006 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17015694

RESUMEN

Identification of candidate genes and their immunological mechanisms that control autoaggressive T cells in inflamed environments, may lead to novel therapies for autoimmune diseases, like type 1 diabetes (T1D). In this study, we used transgenic NOD mice that constitutively express TNF-alpha in their islets from neonatal life (TNF-alpha-NOD) to identify protective alleles that control T1D in the presence of a proinflammatory environment. We show that TNF-alpha-mediated breakdown in T cell tolerance requires recessive NOD alleles. To identify some of these recessive alleles, we crossed TNF-alpha-NOD mice to diabetes-resistant congenic NOD mice having protective alleles at insulin-dependent diabetes (Idd) loci that control spontaneous T1D at either the preinsulitis (Idd3.Idd5) or postinsulitis (Idd9) phases. No protection from TNF-alpha-accelerated T1D was afforded by resistance alleles at Idd3.Idd5. Lack of protection was not at the level of T cell priming, the efficacy of islet-infiltrating APCs to present islet peptides, nor the ability of high levels of CD4+ Foxp3+ T cells to accumulate in the islets. In contrast, protective alleles at Idd9 significantly increased the age at which TNF-alpha-NOD mice developed T1D. Disease delay was associated with a decreased ability of CD8+ T cells to respond to islet Ags presented by islet-infiltrating APCs. Finally, we demonstrate that the protective region on chromosome 4 that controls T1D in TNF-alpha-Idd9 mice is restricted to the Idd9.1 region. These data provide new evidence of the mechanisms by which selective genetic loci control autoimmune diseases in the presence of a strong inflammatory assault.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Cromosomas/inmunología , Diabetes Mellitus Tipo 1/patología , Células Secretoras de Insulina/inmunología , Factor de Necrosis Tumoral alfa/genética , Alelos , Animales , Presentación de Antígeno/inmunología , Diabetes Mellitus Tipo 1/genética , Tolerancia Inmunológica/genética , Inmunidad Innata/genética , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Linfocitos T Citotóxicos/inmunología , Factor de Necrosis Tumoral alfa/inmunología
17.
Int Immunol ; 17(6): 721-8, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15899926

RESUMEN

The immune system has co-evolved with the infectious agents that challenge it, and in response pathogens have developed different mechanisms to subvert host immunity. A wealth of evidence suggests that infections are important components in the development of a functional immune system, and understanding the modulation of the host immune system by pathogens may offer new therapeutic strategies in a non-infectious setting. We investigated how infection with the protozoan parasite Trypanosoma brucei brucei (Tbb) modulates the autoimmune response to recombinant myelin oligodendrocyte glycoprotein (rMOG) in DBA/1 mice. Mice harbouring a Tbb infection did not develop experimental autoimmune encephalomyelitis (EAE) induced by immunization with rMOG in CFA, an animal model for the human autoimmune disease multiple sclerosis. Additionally, mice infected with the parasite at the time of immunization or 1 week later developed less severe EAE than uninfected controls. Protected mice displayed a markedly diminished rMOG-specific proliferation and IFNgamma production in lymph node cells and had correspondingly low titres of serum anti-rMOG IgG. Antigen-presenting cells (APCs) from spleens of Tbb-infected mice presented rMOG less efficiently to rMOG-specific T cells in vitro than did splenic APCs from uninfected mice and could also inhibit antigen-specific proliferation in control in vitro cultures. This suppressive effect is at least in part due to increased release of IL-10. Transfer of splenic APCs from Tbb-infected mice into mice immunized with rMOG-CFA 7 days previously abrogated disease significantly. These findings indicate that infections can prevent autoimmunity and that APCs might be used as immunomodulants.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Glicoproteína Asociada a Mielina/inmunología , Trypanosoma brucei brucei/inmunología , Tripanosomiasis Africana/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/prevención & control , Femenino , Inmunoglobulina G/sangre , Terapia de Inmunosupresión , Interferón gamma/metabolismo , Interleucina-10/inmunología , Interleucina-10/metabolismo , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos DBA , Proteínas de la Mielina , Glicoproteína Asociada a Mielina/administración & dosificación , Glicoproteína Mielina-Oligodendrócito , Bazo
18.
Eur J Immunol ; 33(6): 1539-47, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12778471

RESUMEN

To prevent an organism from developing autoimmunity the body limits the number of autoreactive cells through thymic negative selection and regulates their activity through induction of suppressor T cells. Development of antigen-specific therapies provides an interesting opportunity to imitate the body's own, often effective, method of protection. Our study demonstrates that DBA/1 mice could be protected from experimental autoimmune encephalomyelitis induced through injection of recombinant myelin oligodendrocyte glycoprotein (rMOG) when they were previously immunized intraperitoneally with rMOG adsorbed to aluminium hydroxide. This protection was associated with a decreased IFN-gamma production by rMOG-specific cells, but not a decreased proliferative response. Protection was long lasting, indicating that MOG-alum vaccination might be developed as a prophylactic therapy in multiple sclerosis.


Asunto(s)
Autoantígenos/inmunología , Encefalomielitis Autoinmune Experimental/prevención & control , Glicoproteína Asociada a Mielina/inmunología , Vacunación , Adyuvantes Inmunológicos , Adsorción , Compuestos de Alumbre , Animales , Autoantígenos/administración & dosificación , Encéfalo/inmunología , Encéfalo/patología , Antígenos CD4/genética , Linfocitos T CD4-Positivos/inmunología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Inmunoglobulina G/inmunología , Terapia de Inmunosupresión , Inyecciones Intraperitoneales , Interferón gamma/biosíntesis , Activación de Linfocitos , Ratones , Ratones Endogámicos DBA , Ratones Noqueados , Esclerosis Múltiple , Proteínas de la Mielina , Glicoproteína Asociada a Mielina/administración & dosificación , Glicoproteína Mielina-Oligodendrócito , Ratas , Proteínas Recombinantes/inmunología , Médula Espinal/inmunología , Médula Espinal/patología , Células Th2/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA